1
|
Nataraj BH, Ramesh C, Mallappa RH. Probiotic and postbiotic interference exhibit anti-adhesion effects against clinical methicillin-resistant Staphylococcus aureus (MRSA) and impede MRSA-induced intestinal epithelial hyper-permeability in HT-29 cell line. Microb Pathog 2025; 199:107215. [PMID: 39647539 DOI: 10.1016/j.micpath.2024.107215] [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: 05/15/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 12/10/2024]
Abstract
This study investigates the dynamics of MRSA de-colonization on HT-29 cell line using effective strategies like probiotics and postbiotics. Exploring novel alternatives to combat infections caused by antibiotic-resistant pathogens is an urgent need. Harnessing the antagonistic properties of live probiotics and their heat-killed preparations (postbiotics) to curb the growth of AMR pathogens represents a promising and essential area of contemporary research. This study was designed to evaluate the anti-adhesion properties of indigenous probiotics (Limosilactobacillus fermentum Lf1 and Lactiplantibacillus plantarum A5), as well as standard reference strains (Lacticaseibacillus rhamnosus GG and Lactobacillus acidophilus NCFM), and their heat-killed postbiotic preparations against clinical MRSA isolates (MRSA12/206 and 5/255) on the HT-29 cell line. ATR-FTIR-based functional group characterization of the postbiotic preparations revealed the heat-induced alterations in cell surface molecules and architecture. Both probiotic and postbiotic preparations were non-cytotoxic to HT-29 cells. The probiotic intervention, via protective, competitive, and displacement modes, significantly (p < 0.05) reduced the adhesion of MRSA isolates to HT-29 cells, with the protective and competitive modes showing greater efficacy. In contrast, heat-killed probiotics demonstrated notable anti-MRSA adhesion effects across all three modes (protective, competitive, and displacement). In comparison, heat-killed cells exhibited a superior anti-adhesion capability compared to live cells, likely due to the enhanced accessibility of microbe-associated molecular patterns and adhesion sites following heat treatment. Furthermore, co-treatment of MRSA with probiotic strains substantially (p < 0.05) reduced FITC-dextran transflux across the HT-29 cell monolayer. In conclusion, this study highlights the superior anti-adhesion efficacy of heat-killed postbiotics over live probiotic cells against MRSA isolates. It underscores the further need for pre-clinical and in-vivo investigations to validate the anti-MRSA colonization and gut barrier prophylactic or therapeutic potential of the investigated probiotics and postbiotics. Thus, the present study documents and supports the alternative to antibiotics potential of probiotics and postbiotics.
Collapse
Affiliation(s)
- Basavaprabhu Haranahalli Nataraj
- Molecular Biology Unit, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India; Dairy Bacteriology Section, Southern Regional Station, ICAR-National Dairy Research Institute, Adugodi, 560030, Bengaluru, Karnataka, India.
| | - Chette Ramesh
- Molecular Biology Unit, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India.
| | - Rashmi Hogarehalli Mallappa
- Molecular Biology Unit, Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, 132001, Haryana, India; Dairy Bacteriology Section, Southern Regional Station, ICAR-National Dairy Research Institute, Adugodi, 560030, Bengaluru, Karnataka, India.
| |
Collapse
|
2
|
Szymczak K, Rychłowski M, Zhang L, Nakonieczna J. Harnessing light-activated gallium porphyrins to combat intracellular Staphylococcus aureus using an in vitro keratinocyte infection model. Sci Rep 2025; 15:1295. [PMID: 39779728 PMCID: PMC11711192 DOI: 10.1038/s41598-024-84312-4] [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: 05/24/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
Staphylococcus aureus (S. aureus) can survive inside nonprofessional phagocytes such as keratinocytes, enabling it to evade antibiotics and cause recurrent infections once treatment stops. New antibacterial strategies to eliminate intracellular, multidrug-resistant bacteria are needed. This study used a keratinocyte model infected with methicillin-resistant S. aureus (MRSA) to test light-activated compounds, specifically heme-mimetic gallium (III) porphyrin (Ga3+CHP) and visible light, known as antimicrobial photodynamic inactivation (aPDI), for eliminating intracellular MRSA. Ga3+CHP was found to accumulate more in infected cells, particularly within lysosomal structures where MRSA resides. Flow cytometry and fluorescence microscopy revealed significant colocalization of MRSA and Ga3+CHP. Under aPDI, MRSA showed reduced adhesion to host cells and a 70% reduction in the GFP signal from intracellular bacteria. Additionally, light-activated Ga3+CHP significantly decreased the number of extracellular bacteria, reducing the potential for further infection. This study is the first to analyze aPDI toxicity in real time within an infection model, demonstrating that this method is neither cytotoxic nor phototoxic.
Collapse
Affiliation(s)
- Klaudia Szymczak
- Laboratory of Photobiology and Molecular Diagnostics, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdańsk, Poland
| | - Michał Rychłowski
- Laboratory of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdańsk, Poland
| | - Lei Zhang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), Tianjin University, Tianjin, China
| | - Joanna Nakonieczna
- Laboratory of Photobiology and Molecular Diagnostics, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdańsk, Poland.
| |
Collapse
|
3
|
Maucotel AL, Crepin DM, Faure A, Valour F, Laurent F, Josse J. Pathogenesis of Staphylococcus epidermidis prosthetic joint infections: bacterial adhesion and internalization in osteoblasts, synoviocytes and endothelial cells. J Med Microbiol 2025; 74. [PMID: 39846993 DOI: 10.1099/jmm.0.001959] [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: 01/24/2025] Open
Abstract
Staphylococcus epidermidis is frequently isolated during prosthetic joint infections (PJIs). Unlike Staphylococcus aureus, its internalization and persistence within cells are controversial. We aimed to determine whether internalization is involved in the pathophysiology of S. epidermidis PJIs. Adhesion and internalization of S. epidermidis PJI isolates have been studied using an in vitro model. Despite similar adhesion levels to the S. aureus SH1000 reference strain, S. epidermidis isolates had a low internalization in osteoblasts, synoviocytes and endothelial cells. Internalization of S. epidermidis is strain- and cell-type dependent. Our results do not support S. epidermidis internalization as a key factor in PJIs.
Collapse
Affiliation(s)
- Anne Lise Maucotel
- Institute for Infectious Agents, Hpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
- Centre International de Recherche en Infectiologie, INSERM U1111, CNRS UMR5308, ENS Lyon, Lyon 1 University, Lyon, France
- French National Reference Centre for Staphylococci, Lyon, France
| | - Deborah M Crepin
- Centre International de Recherche en Infectiologie, INSERM U1111, CNRS UMR5308, ENS Lyon, Lyon 1 University, Lyon, France
| | - Allison Faure
- Centre International de Recherche en Infectiologie, INSERM U1111, CNRS UMR5308, ENS Lyon, Lyon 1 University, Lyon, France
| | - Florent Valour
- Centre International de Recherche en Infectiologie, INSERM U1111, CNRS UMR5308, ENS Lyon, Lyon 1 University, Lyon, France
| | - Frédéric Laurent
- Institute for Infectious Agents, Hpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
- Centre International de Recherche en Infectiologie, INSERM U1111, CNRS UMR5308, ENS Lyon, Lyon 1 University, Lyon, France
- French National Reference Centre for Staphylococci, Lyon, France
- Microbiology-Mycology Department, Institut des Sciences Pharmaceutiques et Biologiques de Lyon, Lyon, France
| | - Jérôme Josse
- Centre International de Recherche en Infectiologie, INSERM U1111, CNRS UMR5308, ENS Lyon, Lyon 1 University, Lyon, France
- Microbiology-Mycology Department, Institut des Sciences Pharmaceutiques et Biologiques de Lyon, Lyon, France
| |
Collapse
|
4
|
Zelmer AR, Yang D, Gunn NJ, Solomon LB, Nelson R, Kidd SP, Richter K, Atkins GJ. Osteomyelitis-relevant antibiotics at clinical concentrations show limited effectivity against acute and chronic intracellular S. aureus infections in osteocytes. Antimicrob Agents Chemother 2024; 68:e0080824. [PMID: 39194210 PMCID: PMC11459924 DOI: 10.1128/aac.00808-24] [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] [Received: 06/02/2024] [Accepted: 08/09/2024] [Indexed: 08/29/2024] Open
Abstract
Osteomyelitis caused by Staphylococcus aureus can involve the persistent infection of osteocytes. We sought to determine if current clinically utilized antibiotics were capable of clearing an intracellular osteocyte S. aureus infection. Rifampicin, vancomycin, levofloxacin, ofloxacin, amoxicillin, oxacillin, doxycycline, linezolid, gentamicin, and tigecycline were assessed for their minimum inhibitory concentration (MIC) and minimum bactericidal concentrations against 12 S. aureus strains, at pH 5.0 and 7.2 to mimic lysosomal and cytoplasmic environments, respectively. Those antibiotics whose bone estimated achievable concentration was commonly above their respective MIC for the strains tested were further assayed in a human osteocyte infection model under acute and chronic conditions. Osteocyte-like cells were treated at 1×, 4×, and 10× the MIC for 1 and 7 days following infection (acute model), or at 15 and 21 days of infection (chronic model). The intracellular effectivity of each antibiotic was measured in terms of CFU reduction, small colony variant formation, and bacterial mRNA expression change. Only rifampicin, levofloxacin, and linezolid reduced intracellular CFU numbers significantly in the acute model. Consistent with the transition to a non-culturable state, few if any CFU could be recovered from the chronic model. However, no treatment in either model reduced the quantity of bacterial mRNA or prevented non-culturable bacteria from returning to a culturable state. These findings indicate that S. aureus adapts phenotypically during intracellular infection of osteocytes, adopting a reversible quiescent state that is protected against antibiotics, even at 10× their MIC. Thus, new therapeutic approaches are necessary to cure S. aureus intracellular infections in osteomyelitis.
Collapse
Affiliation(s)
- Anja R. Zelmer
- Center for Orthopedic and Trauma Research, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | - Dongqing Yang
- Center for Orthopedic and Trauma Research, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | - Nicholas J. Gunn
- Center for Orthopedic and Trauma Research, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | - L. Bogdan Solomon
- Center for Orthopedic and Trauma Research, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
- Department of Orthopedics and Trauma, Royal Adelaide Hospital, Adelaide, Australia
| | - Renjy Nelson
- Department of Infectious Diseases, Central Adelaide Local Health Network, Adelaide, Australia
| | - Stephen P. Kidd
- Australian Center for Antimicrobial Resistance Ecology, University of Adelaide, Adelaide, Australia
- Research Center for Infectious Disease, School of Biological Science, University of Adelaide, Adelaide, Australia
| | - Katharina Richter
- Department of Surgery, Richter Lab, Basil Hetzel Institute for Translational Health Research, University of Adelaide, Adelaide, Australia
- Institute for Photonics and Advanced Sensing, University of Adelaide, Adelaide, Australia
| | - Gerald J. Atkins
- Center for Orthopedic and Trauma Research, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| |
Collapse
|
5
|
Yuan K, Yang Y, Lin Y, Zhou F, Huang K, Yang S, Kong W, Li F, Kan T, Wang Y, Cheng C, Liang Y, Chang H, Huang J, Ao H, Yu Z, Li H, Liu Y, Tang T. Targeting Bacteria-Induced Ferroptosis of Bone Marrow Mesenchymal Stem Cells to Promote the Repair of Infected Bone Defects. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404453. [PMID: 39166412 PMCID: PMC11497072 DOI: 10.1002/advs.202404453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/30/2024] [Indexed: 08/22/2024]
Abstract
The specific mechanisms underlying bacteria-triggered cell death and osteogenic dysfunction in host bone marrow mesenchymal stem cells (BMSCs) remain unclear, posing a significant challenge to the repair of infected bone defects. This study identifies ferroptosis as the predominant cause of BMSCs death in the infected bone microenvironment. Mechanistically, the bacteria-induced activation of the innate immune response in BMSCs leads to upregulation and phosphorylation of interferon regulatory factor 7 (IRF7), thus facilitating IRF7-dependent ferroptosis of BMSCs through the transcriptional upregulation of acyl-coenzyme A synthetase long-chain family member 4 (ACSL4). Moreover, it is found that intervening in ferroptosis can partially rescue cell injuries and osteogenic dysfunction. Based on these findings, a hydrogel composite 3D-printed scaffold is designed with reactive oxygen species (ROS)-responsive release of antibacterial quaternized chitosan and sustained delivery of the ferroptosis inhibitor Ferrostatin-1 (Fer-1), capable of eradicating pathogens and promoting bone regeneration in a rat model of infected bone defects. Together, this study suggests that ferroptosis of BMSCs is a promising therapeutic target for infected bone defect repair.
Collapse
Affiliation(s)
- Kai Yuan
- Shanghai Key Laboratory of Orthopaedic ImplantsDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
| | - Yiqi Yang
- Department of OrthopedicsThe First Affiliated HospitalZhejiang University School of Medicine79 Qingchun RdHangzhou310003P. R. China
| | - Yixuan Lin
- Shanghai Key Laboratory of Orthopaedic ImplantsDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
| | - Feng Zhou
- Department of Orthopaedic SurgeryThe First Affiliated Hospital of Soochow UniversityNo. 899 Ping Hai RoadSuzhouJiangsu215006P. R. China
| | - Kai Huang
- Shanghai Key Laboratory of Orthopaedic ImplantsDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
| | - Shengbing Yang
- Shanghai Key Laboratory of Orthopaedic ImplantsDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
| | - Weiqing Kong
- Department of Orthopaedic SurgeryXuzhou Central HospitalXuzhou Clinical School of Xuzhou Medical University199 Jiefang South RoadXuzhou221009P. R. China
| | - Fupeng Li
- Shanghai Key Laboratory of Orthopaedic ImplantsDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
| | - Tianyou Kan
- Shanghai Key Laboratory of Orthopaedic ImplantsDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
| | - Yao Wang
- Shanghai Key Laboratory of Orthopaedic ImplantsDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
| | - Caiqi Cheng
- Shanghai Key Laboratory of Orthopaedic ImplantsDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
| | - Yakun Liang
- Shanghai Institute of Precision MedicineShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200125P. R. China
| | - Haishuang Chang
- Shanghai Institute of Precision MedicineShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200125P. R. China
| | - Jie Huang
- Shanghai Institute of Precision MedicineShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200125P. R. China
| | - Haiyong Ao
- Jiangxi Key Laboratory of Nanobiomaterials & School of Materials Science and EngineeringEast China Jiaotong UniversityNanchang330000P. R. China
| | - Zhifeng Yu
- Shanghai Key Laboratory of Orthopaedic ImplantsDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
| | - Hanjun Li
- State Key Laboratory of Systems Medicine for CancerRenji‐Med X Clinical Stem Cell Research CenterRen Ji HospitalShanghai Jiao Tong University School of Medicine160 Pujian RoadShanghai200127P. R. China
| | - Yihao Liu
- Shanghai Key Laboratory of Orthopaedic ImplantsDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
| | - Tingting Tang
- Shanghai Key Laboratory of Orthopaedic ImplantsDepartment of Orthopaedic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
| |
Collapse
|
6
|
Dashtbin S, Razavi S, Ganjali Koli M, Barneh F, Ekhtiari-Sadegh S, Akbari R, Irajian G, Pooshang Bagheri K. Intracellular bactericidal activity and action mechanism of MDP1 antimicrobial peptide against VRSA and MRSA in human endothelial cells. Front Microbiol 2024; 15:1416995. [PMID: 39252832 PMCID: PMC11381295 DOI: 10.3389/fmicb.2024.1416995] [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: 04/13/2024] [Accepted: 08/05/2024] [Indexed: 09/11/2024] Open
Abstract
Introduction Staphylococcus aureus is a prominent cause of postoperative infections, often persisting within host cells, leading to chronic infections. Conventional antibiotics struggle to eliminate intracellular S. aureus due to poor cell penetration. Antimicrobial peptides are a new hope for tackling intracellular bacteria. Accordingly, this study examines the antimicrobial peptide MDP1, derived from melittin, for its efficacy against intracellular S. aureus. Methods In this study, the physiochemical properties (Prediction of three-dimensional structure, circular dichroism and helical wheel projection analysis) were investigated. Extracellular antibacterial activity and cytotoxicity of MDP1 were also assessed. The mechanism of interaction of MDP1 with S. aureus was evaluated by molecular dynamic simulation, atomic force and confocal microscopy. Bacterial internalization into an endothelial cell model was confirmed through culture and transmission electron microscopy. The effect of the peptide on intracellular bacteria was investigated by culture and epi-fluorescence microscopy. Results and discussion 3D structural prediction proved the conformation of MDP1 as an α-helix peptide. Helical-wheel projection analysis indicated the proper orientation of hydrophobic amino acid residues for membrane interaction. CD spectroscopy of MDP1 showed that MDP1 in SDS 10 and 30 mM adopted 87 and 91% helical conformation. Atomic force and confocal microscopy assessments as well as molecular dynamics studies revealed the peptide-bacterial membrane interaction. MDP1, at the concentration of 0.32 μg mL-1, demonstrated a fold reduction of 21.7 ± 1.8, 1.7 ± 0.2, and 7.3 ± 0.8 in intracellular bacterial load for ATCC, VRSA, and MRSA, respectively. Molecular dynamics results demonstrate a preferential interaction of MDP1 with POPG/POPE membranes, primarily driven by electrostatic forces and hydrogen bonding. In POPC systems, two out of four MDP1 interacted effectively, while all four MDP1 engaged with POPG/POPE membranes. Gathering all data together, MDP1 is efficacious in the reduction of intracellular VRSA and MRSA proved by culture and epi-fluorescent microscopy although further studies should be performed to increase the intracellular activity of MDP1.
Collapse
Affiliation(s)
- Shirin Dashtbin
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shabnam Razavi
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mokhtar Ganjali Koli
- Department of Chemistry, University of Kurdistan, Sanandaj, Iran
- Computational Chemistry Laboratory, Kask Afrand Exire Ltd., Sanandaj, Iran
| | - Farnoosh Barneh
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Sarvenaz Ekhtiari-Sadegh
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Reza Akbari
- Department of Microbiology and Virology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Gholamreza Irajian
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Microbiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Kamran Pooshang Bagheri
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
7
|
Mills EL, Avila YI, Beasock D, Radwan Y, Suptela SR, Marriott I, Afonin KA, Johnson MB. Immunostimulatory nucleic acid nanoparticles (NANPs) augment protective osteoblast and osteoclast type I interferon responses to Staphylococcus aureus. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 60:102762. [PMID: 38866196 PMCID: PMC11297679 DOI: 10.1016/j.nano.2024.102762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 06/14/2024]
Abstract
Recalcitrant staphylococcal osteomyelitis may be due, in part, to the ability of Staphylococcus aureus to invade bone cells. However, osteoclasts and osteoblasts are now recognized to shape host responses to bacterial infection and we have recently described their ability to produce IFN-β following S. aureus infection and limit intracellular bacterial survival/propagation. Here, we have investigated the ability of novel, rationally designed, nucleic acid nanoparticles (NANPs) to induce the production of immune mediators, including IFN-β, following introduction into bone cells. We demonstrate the successful delivery of representative NANPs into osteoblasts and osteoclasts via endosomal trafficking when complexed with lipid-based carriers. Their delivery was found to differentially induce immune responses according to their composition and architecture via discrete cytosolic pattern recognition receptors. Finally, the utility of this nanoparticle technology was supported by the demonstration that immunostimulatory NANPs augment IFN-β production by S. aureus infected bone cells and reduce intracellular bacterial burden.
Collapse
Affiliation(s)
- Erin L Mills
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA, 28223.
| | - Yelixza I Avila
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, USA, 28223.
| | - Damian Beasock
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, USA, 28223
| | - Yasmine Radwan
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, USA, 28223.
| | - Samantha R Suptela
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA, 28223.
| | - Ian Marriott
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA, 28223.
| | - Kirill A Afonin
- Department of Chemistry, University of North Carolina at Charlotte, Charlotte, NC, USA, 28223.
| | - M Brittany Johnson
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, USA, 28223.
| |
Collapse
|
8
|
Long DR, Holmes EA, Lo HY, Penewit K, Almazan J, Hodgson T, Berger NF, Bishop ZH, Lewis JD, Waalkes A, Wolter DJ, Salipante SJ. Clinical and in vitro models identify distinct adaptations enhancing Staphylococcus aureus pathogenesis in human macrophages. PLoS Pathog 2024; 20:e1012394. [PMID: 38991026 PMCID: PMC11265673 DOI: 10.1371/journal.ppat.1012394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 07/23/2024] [Accepted: 07/04/2024] [Indexed: 07/13/2024] Open
Abstract
Staphylococcus aureus is a facultative intracellular pathogen of human macrophages, which facilitates chronic infection. The genotypes, pathways, and mutations influencing that phenotype remain incompletely explored. Here, we used two distinct strategies to ascertain S. aureus gene mutations affecting pathogenesis in macrophages. First, we analyzed isolates collected serially from chronic cystic fibrosis (CF) respiratory infections. We found that S. aureus strains evolved greater macrophage invasion capacity during chronic human infection. Bacterial genome-wide association studies (GWAS) identified 127 candidate genes for which mutation was significantly associated with macrophage pathogenesis in vivo. In parallel, we passaged laboratory S. aureus strains in vitro to select for increased infection of human THP-1 derived macrophages, which identified 15 candidate genes by whole-genome sequencing. Functional validation of candidate genes using isogenic transposon mutant knockouts and CRISPR interference (CRISPRi) knockdowns confirmed virulence contributions from 37 of 39 tested genes (95%) implicated by in vivo studies and 7 of 10 genes (70%) ascertained from in vitro selection, with one gene in common to the two strategies. Validated genes included 17 known virulence factors (39%) and 27 newly identified by our study (61%), some encoding functions not previously associated with macrophage pathogenesis. Most genes (80%) positively impacted macrophage invasion when disrupted, consistent with the phenotype readily arising from loss-of-function mutations in vivo. This work reveals genes and mechanisms that contribute to S. aureus infection of macrophages, highlights differences in mutations underlying convergent phenotypes arising from in vivo and in vitro systems, and supports the relevance of S. aureus macrophage pathogenesis during chronic respiratory infection in CF. Additional studies will be needed to illuminate the exact mechanisms by which implicated mutations affect their phenotypes.
Collapse
Affiliation(s)
- Dustin R. Long
- Division of Critical Care Medicine, Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Elizabeth A. Holmes
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Hsin-Yu Lo
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Kelsi Penewit
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Jared Almazan
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Taylor Hodgson
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Nova F. Berger
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Zoe H. Bishop
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Janessa D. Lewis
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Adam Waalkes
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Daniel J. Wolter
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Stephen J. Salipante
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, United States of America
| |
Collapse
|
9
|
Xu H, Wang X, Zhang Z, Hu J, Yu Y, Wang J, Liu Y, Liu J. Staphylococcus aureus promotes its intracellular survival by inhibiting Rab11-Rab11FIP4-mediated vesicle trafficking. Vet Microbiol 2024; 293:110091. [PMID: 38626624 DOI: 10.1016/j.vetmic.2024.110091] [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: 09/07/2023] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/18/2024]
Abstract
Mastitis in dairy cows is mainly caused by bacteria, in which Staphylococcus aureus appears frequently. Epithelial cells, as a major physical barrier of mammary gland, play an important role in preventing mastitis in dairy cows. Our previous study reported that Rab11fip4 (an effector of Rab11) was significantly changed in response to stimulation by S. aureus. So, in this study, the role of Rab11A in phagocytosis of bovine mammary epithelial cells (MAC-T) against S. aureus was evaluated. First, changes of Rab11A and Rab11fip4 were analyzed in response to S. aureus by immunofluorescence and western blotting. Subsequently, the effects of Rab11A and Rab11fip4 on proliferation of S. aureus, as well as formation and function of late endosomes (LEs) and lysosomes (LYSs) were investigated. The results showed that, after infection, Rab11A and Rab11fip4 were recruited to phagosomes containing S. aureus. Rab11A promoted bacterial clearance and rescues the destruction of LEs and LYSs by S. aureus, whereas Rab11fip4 did the opposite. These findings provide new insights into phagocytosis and control of S. aureus in host cells, thus lay the foundation to elucidate the pathogenesis of S. aureus in bovine mastitis.
Collapse
Affiliation(s)
- Huiling Xu
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong 271018, China
| | - Xiaozhou Wang
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong 271018, China
| | - Zhizhong Zhang
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong 271018, China
| | - Jiaqing Hu
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong 271018, China
| | - Yongtao Yu
- School of Animal Science and Technology, Ningxia University, Yinchuan 750000, China
| | - Jiandong Wang
- Institute of Animal Science, Ningxia Academy of Agricultural and Forestry Sciences, Yinchuan, Ningxia 750002, China
| | - Yongxia Liu
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong 271018, China; Research Center for Animal Disease Control Engineering, Shandong Agricultural University, Tai`an, Shandong 271018, China.
| | - Jianzhu Liu
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong 271018, China.
| |
Collapse
|
10
|
Li J, Zheng H, Leung SSY. Investigating the effectiveness of liposome-bacteriophage nanocomplex in killing Staphylococcus aureus using epithelial cell coculture models. Int J Pharm 2024; 657:124146. [PMID: 38657716 DOI: 10.1016/j.ijpharm.2024.124146] [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: 12/19/2023] [Revised: 04/11/2024] [Accepted: 04/20/2024] [Indexed: 04/26/2024]
Abstract
Host cell invasion with strong antibiotics evading is a major feature of respiratory Staphylococcus aureus infections with severe recurrence. Bacteriophage (phage) therapy and design of liposomal phage to target intracellular pathogens have been described recently. The practicality for pulmonary delivery of liposomal phage, and how formulation compositions affecting the aerosolization and intracellular bacterial killing remain unexplored. In the present study, three commonly used phospholipids (SPC, EPC, and HSPC) were selected to investigate their ability for phage K nebulization and intracellular therapy in the form of liposome-phage nanocomplexes. The three lipid nanocarriers showed protection on phage K upon mesh nebulization and the pulmonary deposition efficiency was influenced by the lipid used. Moreover, the intracellular bacterial killing was strongly depended on the lipid types, where EPC-phage exhibited the best killing performance with no relapsing. Phage K with the aid of EPC liposomes was also observed to manage the tissue infection in a 3D spheroid model more effectively than other groups. Altogether, this novel EPC liposome-phage nanocomplex can be a promising formulation approach that enables inhalable phage to manage respiratory infections caused by bacteria strongly associated with human epithelial cells.
Collapse
Affiliation(s)
- Jiaqi Li
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Huangliang Zheng
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong
| | | |
Collapse
|
11
|
Doğan E, Sydow K, Heiden SE, Eger E, Wassilew G, Proctor RA, Bohnert JA, Idelevich EA, Schaufler K, Becker K. Klebsiella pneumoniae exhibiting a phenotypic hyper-splitting phenomenon including the formation of small colony variants. Front Cell Infect Microbiol 2024; 14:1372704. [PMID: 38601740 PMCID: PMC11004228 DOI: 10.3389/fcimb.2024.1372704] [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: 01/18/2024] [Accepted: 03/07/2024] [Indexed: 04/12/2024] Open
Abstract
In this study, we characterized a Klebsiella pneumoniae strain in a patient with shrapnel hip injury, which resulted in multiple phenotypic changes, including the formation of a small colony variant (SCV) phenotype. Although already described since the 1960s, there is little knowledge about SCV phenotypes in Enterobacteriaceae. The formation of SCVs has been recognized as a bacterial strategy to evade host immune responses and compromise the efficacy of antimicrobial therapies, leading to persistent and recurrent courses of infections. In this case, 14 isolates with different resisto- and morpho-types were distinguished from the patient's urine and tissue samples. Whole genome sequencing revealed that all isolates were clonally identical belonging to the K. pneumoniae high-risk sequence type 147. Subculturing the SCV colonies consistently resulted in the reappearance of the initial SCV phenotype and three stable normal-sized phenotypes with distinct morphological characteristics. Additionally, an increase in resistance was observed over time in isolates that shared the same colony appearance. Our findings highlight the complexity of bacterial behavior by revealing a case of phenotypic "hyper-splitting" in a K. pneumoniae SCV and its potential clinical significance.
Collapse
Affiliation(s)
- Eyüp Doğan
- Friedrich Loeffler-Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Katharina Sydow
- Department of Epidemiology and Ecology of Antimicrobial Resistance, Helmholtz Institute for One Health, Helmholtz Centre for Infection Research Helmholtz Center for Infection Research (HZI), Greifswald, Germany
| | - Stefan E. Heiden
- Department of Epidemiology and Ecology of Antimicrobial Resistance, Helmholtz Institute for One Health, Helmholtz Centre for Infection Research Helmholtz Center for Infection Research (HZI), Greifswald, Germany
| | - Elias Eger
- Department of Epidemiology and Ecology of Antimicrobial Resistance, Helmholtz Institute for One Health, Helmholtz Centre for Infection Research Helmholtz Center for Infection Research (HZI), Greifswald, Germany
| | - Georgi Wassilew
- Center for Orthopedics, Trauma Surgery and Rehabilitation Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Richard A. Proctor
- Departments of Medical Microbiology/Immunology and Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Jürgen A. Bohnert
- Friedrich Loeffler-Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Evgeny A. Idelevich
- Friedrich Loeffler-Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Katharina Schaufler
- Department of Epidemiology and Ecology of Antimicrobial Resistance, Helmholtz Institute for One Health, Helmholtz Centre for Infection Research Helmholtz Center for Infection Research (HZI), Greifswald, Germany
- University Medicine Greifswald, Greifswald, Germany
| | - Karsten Becker
- Friedrich Loeffler-Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
12
|
Liu A, Garrett S, Hong W, Zhang J. Staphylococcus aureus Infections and Human Intestinal Microbiota. Pathogens 2024; 13:276. [PMID: 38668232 PMCID: PMC11053856 DOI: 10.3390/pathogens13040276] [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: 02/16/2024] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 04/29/2024] Open
Abstract
Staphylococcus aureus (S. aureus) is a common pathogen that can cause many human diseases, such as skin infection, food poisoning, endocarditis, and sepsis. These diseases can be minor infections or life-threatening, requiring complex medical management resulting in substantial healthcare costs. Meanwhile, as the critically ignored "organ," the intestinal microbiome greatly impacts physiological health, not only in gastrointestinal diseases but also in disorders beyond the gut. However, the correlation between S. aureus infection and intestinal microbial homeostasis is largely unknown. Here, we summarized the recent progress in understanding S. aureus infections and their interactions with the microbiome in the intestine. These summarizations will help us understand the mechanisms behind these infections and crosstalk and the challenges we are facing now, which could contribute to preventing S. aureus infections, effective treatment investigation, and vaccine development.
Collapse
Affiliation(s)
- Aotong Liu
- Department of Pharmacology & Regenerative Medicine, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA;
| | - Shari Garrett
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA;
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Wanqing Hong
- Faculty of Science, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
- School of Chemistry & Chemical Engineering and Materials Sciences, Shandong Normal University, Jinan 250061, China
| | - Jilei Zhang
- Department of Pharmacology & Regenerative Medicine, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA;
| |
Collapse
|
13
|
Mascary JB, Bordeau V, Nicolas I, Verdier MC, Rocheteau P, Cattoir V. Intracellular activity and in vivo efficacy in a mouse model of septic arthritis of the novel pseudopeptide Pep16 against Staphylococcus aureus clinical isolates. JAC Antimicrob Resist 2024; 6:dlae025. [PMID: 38410249 PMCID: PMC10895697 DOI: 10.1093/jacamr/dlae025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 02/02/2024] [Indexed: 02/28/2024] Open
Abstract
Objectives Assessing the therapeutic potential of a novel antimicrobial pseudopeptide, Pep16, both in vitro and in vivo for the treatment of septic arthritis caused by Staphylococcus aureus. Methods Seven clinical isolates of S. aureus (two MRSA and five MSSA) were studied. MICs of Pep16 and comparators (vancomycin, teicoplanin, daptomycin and levofloxacin) were determined through the broth microdilution method. The intracellular activity of Pep16 and levofloxacin was assessed in two models of infection using non-professional (osteoblasts MG-63) or professional (macrophages THP-1) phagocytic cells. A mouse model of septic arthritis was used to evaluate the in vivo efficacy of Pep16 and vancomycin. A preliminary pharmacokinetic (PK) analysis was performed by measuring plasma concentrations using LC-MS/MS following a single subcutaneous injection of Pep16 (10 mg/kg). Results MICs of Pep16 were consistently at 8 mg/L for all clinical isolates of S. aureus (2- to 32-fold higher to those of comparators) while MBC/MIC ratios confirmed its bactericidal activity. Both Pep16 and levofloxacin (when used at 2 × MIC) significantly reduced the bacterial load of all tested isolates (two MSSA and two MRSA) within both osteoblasts and macrophages. In MSSA-infected mice, Pep16 demonstrated a significant (∼10-fold) reduction on bacterial loads in knee joints. PK analysis following a single subcutaneous administration of Pep16 revealed a gradual increase in plasma concentrations, reaching a peak of 5.6 mg/L at 12 h. Conclusions Pep16 is a promising option for the treatment of septic arthritis due to S. aureus, particularly owing to its robust intracellular activity.
Collapse
Affiliation(s)
- Jean-Baptiste Mascary
- Inserm U1230 BRM (Bacterial RNAs and Medicine), Université de Rennes, Rennes, France
- SAS Olgram, Bréhan, France
| | - Valérie Bordeau
- Inserm U1230 BRM (Bacterial RNAs and Medicine), Université de Rennes, Rennes, France
| | | | | | | | - Vincent Cattoir
- CHU de Rennes, Service de Bactériologie-Hygiène hospitalière, 2 rue Henri Le Guilloux, 35033 Rennes, France
- CNR de la Résistance aux Antibiotiques (laboratoire associé 'Entérocoques'), CHU de Rennes, Rennes, France
| |
Collapse
|
14
|
Goormaghtigh F, Van Bambeke F. Understanding Staphylococcus aureus internalisation and induction of antimicrobial tolerance. Expert Rev Anti Infect Ther 2024; 22:87-101. [PMID: 38180805 DOI: 10.1080/14787210.2024.2303018] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/04/2024] [Indexed: 01/07/2024]
Abstract
INTRODUCTION Staphylococcus aureus, a human commensal, is also one of the most common and serious pathogens for humans. In recent years, its capacity to survive and replicate in phagocytic and non-phagocytic cells has been largely demonstrated. In these intracellular niches, bacteria are shielded from the immune response and antibiotics, turning host cells into long-term infectious reservoirs. Moreover, neutrophils carry intracellular bacteria in the bloodstream, leading to systemic spreading of the disease. Despite the serious threat posed by intracellular S. aureus to human health, the molecular mechanisms behind its intracellular survival and subsequent antibiotic treatment failure remain elusive. AREA COVERED We give an overview of the killing mechanisms of phagocytes and of the impressive arsenal of virulence factors, toxins and stress responses deployed by S. aureus as a response. We then discuss the different barriers to antibiotic activity in this intracellular niche and finally describe innovative strategies to target intracellular persisting reservoirs. EXPERT OPINION Intracellular niches represent a challenge in terms of diagnostic and treatment. Further research using ad-hoc in-vivo models and single cell approaches are needed to better understand the molecular mechanisms underlying intracellular survival and tolerance to antibiotics in order to identify strategies to eliminate these persistent bacteria.
Collapse
Affiliation(s)
- Frédéric Goormaghtigh
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Françoise Van Bambeke
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
15
|
Youssouf N, Martin M, Bischoff M, Soubeyran P, Gannoun-Zaki L, Molle V. The secreted tyrosine phosphatase PtpA promotes Staphylococcus aureus survival in RAW 264.7 macrophages through decrease of the SUMOylation host response. Microbiol Spectr 2023; 11:e0281323. [PMID: 37819153 PMCID: PMC10714793 DOI: 10.1128/spectrum.02813-23] [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: 07/11/2023] [Accepted: 09/04/2023] [Indexed: 10/13/2023] Open
Abstract
IMPORTANCE Staphylococcus aureus uses numerous strategies to survive and persist in the intracellular environment of professional phagocytes, including modulation of the SUMOylation process. This study aims to understand how S. aureus alters host SUMOylation to enhance its intracellular survival in professional phagocytes. Our results indicate that S. aureus strain Newman utilizes PtpA-driven phosphorylation to decrease the amount of SUMOylated proteins in murine macrophages to facilitate its survival in this immune cell type.
Collapse
Affiliation(s)
- Nadhuma Youssouf
- VBIC, INSERM U1047, Université de Montpellier, Montpellier, France
| | - Marianne Martin
- VBIC, INSERM U1047, Université de Montpellier, Montpellier, France
| | - Markus Bischoff
- Institute for Medical Microbiology and Hygiene, Saarland University, Homburg, Saarland, Germany
| | - Philippe Soubeyran
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR, Aix-Marseille, Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | | | - Virginie Molle
- VBIC, INSERM U1047, Université de Montpellier, Montpellier, France
| |
Collapse
|
16
|
Friot A, Djebali S, Valsesia S, Parroche P, Dubois M, Baude J, Vandenesch F, Marvel J, Leverrier Y. Antigen specific activation of cytotoxic CD8 + T cells by Staphylococcus aureus infected dendritic cells. Front Cell Infect Microbiol 2023; 13:1245299. [PMID: 37953797 PMCID: PMC10639145 DOI: 10.3389/fcimb.2023.1245299] [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: 06/23/2023] [Accepted: 09/22/2023] [Indexed: 11/14/2023] Open
Abstract
Staphylococcus aureus (S. aureus) is a pathogen associated with a wide variety of diseases, from minor to life-threatening infections. Antibiotic-resistant strains have emerged, leading to increasing concern about the control of S. aureus infections. The development of vaccines may be one way to overcome these resistant strains. However, S. aureus ability to internalize into cells - and thus to form a reservoir escaping humoral immunity - is a challenge for vaccine development. A role of T cells in the elimination of persistent S. aureus has been established in mice but it remains to be established if CD8+ T cells could display a cytotoxic activity against S. aureus infected cells. We examined in vitro the ability of CD8+ T cells to recognize and kill dendritic cells infected with S. aureus. We first evidenced that both primary mouse dendritic cells and DC2.4 cell line can be infected with S. aureus. We then generated a strain of S. aureus expressing a model CD8 epitope and transgenic F5 CD8+ T cells recognizing this model epitope were used as reporter T cells. In response to S. aureus-infected dendritic cells, F5 CD8+ T cells produced IFN-γ in an antigen-specific manner and displayed an increased ability to kill infected cells. Altogether, these results demonstrate that cells infected by S. aureus display bacteria-derived epitopes at their surface that are recognized by CD8+ T cells. This paves the way for the development of CD8+ T cell-based therapies against S. aureus.
Collapse
|
17
|
Lo HY, Long DR, Holmes EA, Penewit K, Hodgson T, Lewis JD, Waalkes A, Salipante SJ. Transposon sequencing identifies genes impacting Staphylococcus aureus invasion in a human macrophage model. Infect Immun 2023; 91:e0022823. [PMID: 37676013 PMCID: PMC10580828 DOI: 10.1128/iai.00228-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 07/13/2023] [Indexed: 09/08/2023] Open
Abstract
Staphylococcus aureus is a facultative intracellular pathogen in many host cell types, facilitating its persistence in chronic infections. The genes contributing to intracellular pathogenesis have not yet been fully enumerated. Here, we cataloged genes influencing S. aureus invasion and survival within human THP-1 derived macrophages using two laboratory strains (ATCC2913 and JE2). We developed an in vitro transposition method to produce highly saturated transposon mutant libraries in S. aureus and performed transposon insertion sequencing (Tn-Seq) to identify candidate genes with significantly altered abundance following macrophage invasion. While some significant genes were strain-specific, 108 were identified as common across both S. aureus strains, with most (n = 106) being required for optimal macrophage infection. We used CRISPR interference (CRISPRi) to functionally validate phenotypic contributions for a subset of genes. Of the 20 genes passing validation, seven had previously identified roles in S. aureus virulence, and 13 were newly implicated. Validated genes frequently evidenced strain-specific effects, yielding opposing phenotypes when knocked down in the alternative strain. Genomic analysis of de novo mutations occurring in groups (n = 237) of clonally related S. aureus isolates from the airways of chronically infected individuals with cystic fibrosis (CF) revealed significantly greater in vivo purifying selection in conditionally essential candidate genes than those not associated with macrophage invasion. This study implicates a core set of genes necessary to support macrophage invasion by S. aureus, highlights strain-specific differences in phenotypic effects of effector genes, and provides evidence for selection of candidate genes identified by Tn-Seq analyses during chronic airway infection in CF patients in vivo.
Collapse
Affiliation(s)
- Hsin-Yu Lo
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Dustin R. Long
- Division of Critical Care Medicine, Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Elizbeth A. Holmes
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Kelsi Penewit
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Taylor Hodgson
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Janessa D. Lewis
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Adam Waalkes
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Stephen J. Salipante
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
18
|
Williams P, Hill P, Bonev B, Chan WC. Quorum-sensing, intra- and inter-species competition in the staphylococci. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001381. [PMID: 37578829 PMCID: PMC10482373 DOI: 10.1099/mic.0.001381] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/31/2023] [Indexed: 08/15/2023]
Abstract
In Gram-positive bacteria such as Staphylococcus aureus and the coagulase-negative staphylococci (CoNS), the accessory gene regulator (agr) is a highly conserved but polymorphic quorum-sensing system involved in colonization, virulence and biofilm development. Signalling via agr depends on the interaction of an autoinducing peptide (AIP) with AgrC, a transmembrane sensor kinase that, once phosphorylated activates the response regulator AgrA. This in turn autoinduces AIP biosynthesis and drives target gene expression directly via AgrA or via the post-transcriptional regulator, RNAIII. In this review we describe the molecular mechanisms underlying the agr-mediated generation of, and response to, AIPs and the molecular basis of AIP-dependent activation and inhibition of AgrC. How the environment impacts on agr functionality is considered and the consequences of agr dysfunction for infection explored. We also discuss the concept of AIP-driven competitive interference between S. aureus and the CoNS and its anti-infective potential.
Collapse
Affiliation(s)
- Paul Williams
- Biodiscovery Institute and School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Phil Hill
- School of Biosciences, Sutton Bonington Campus, University of Nottingham, Loughborough, LE12 5RD, UK
| | - Boyan Bonev
- Biodiscovery Institute and School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Weng C. Chan
- School of Pharmacy, Biodiscovery Institute, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| |
Collapse
|
19
|
Youn C, Pontaza C, Wang Y, Dikeman DA, Joyce DP, Alphonse MP, Wu MJ, Nolan SJ, Anany MA, Ahmadi M, Young J, Tocaj A, Garza LA, Wajant H, Miller LS, Archer NK. Neutrophil-intrinsic TNF receptor signaling orchestrates host defense against Staphylococcus aureus. SCIENCE ADVANCES 2023; 9:eadf8748. [PMID: 37327341 PMCID: PMC10275602 DOI: 10.1126/sciadv.adf8748] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 05/11/2023] [Indexed: 06/18/2023]
Abstract
Staphylococcus aureus is the leading cause of skin and soft tissue infections and is a major health burden due to the emergence of antibiotic-resistant strains. To address the unmet need of alternative treatments to antibiotics, a better understanding of the protective immune mechanisms against S. aureus skin infection is warranted. Here, we report that tumor necrosis factor (TNF) promoted protection against S. aureus in the skin, which was mediated by bone marrow-derived immune cells. Furthermore, neutrophil-intrinsic TNF receptor (TNFR) signaling directed immunity against S. aureus skin infections. Mechanistically, TNFR1 promoted neutrophil recruitment to the skin, whereas TNFR2 prevented systemic bacterial dissemination and directed neutrophil antimicrobial functions. Treatment with a TNFR2 agonist showed therapeutic efficacy against S. aureus and Pseudomonas aeruginosa skin infections, which involved increased neutrophil extracellular trap formation. Our findings revealed nonredundant roles for TNFR1 and TNFR2 in neutrophils for immunity against S. aureus and can be therapeutically targeted for protection against bacterial skin infections.
Collapse
Affiliation(s)
- Christine Youn
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Cristina Pontaza
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Yu Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Dustin A. Dikeman
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Daniel P. Joyce
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Martin P. Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Meng-Jen Wu
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Sabrina J. Nolan
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Mohamed A. Anany
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg 97080, Germany
- Department of Microbial Biotechnology, Institute of Biotechnology, National Research Center, El Buhouth Street, Dokki, 12622 Giza, Egypt
| | - Michael Ahmadi
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Jeremy Young
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Aron Tocaj
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Luis A. Garza
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg 97080, Germany
| | - Lloyd S. Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| | - Nathan K. Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD 21287, USA
| |
Collapse
|
20
|
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
|
21
|
Hachani A, Giulieri SG, Guérillot R, Walsh CJ, Herisse M, Soe YM, Baines SL, Thomas DR, Cheung SD, Hayes AS, Cho E, Newton HJ, Pidot S, Massey RC, Howden BP, Stinear TP. A high-throughput cytotoxicity screening platform reveals agr-independent mutations in bacteraemia-associated Staphylococcus aureus that promote intracellular persistence. eLife 2023; 12:e84778. [PMID: 37289634 PMCID: PMC10259494 DOI: 10.7554/elife.84778] [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: 11/08/2022] [Accepted: 05/23/2023] [Indexed: 06/10/2023] Open
Abstract
Staphylococcus aureus infections are associated with high mortality rates. Often considered an extracellular pathogen, S. aureus can persist and replicate within host cells, evading immune responses, and causing host cell death. Classical methods for assessing S. aureus cytotoxicity are limited by testing culture supernatants and endpoint measurements that do not capture the phenotypic diversity of intracellular bacteria. Using a well-established epithelial cell line model, we have developed a platform called InToxSa (intracellular toxicity of S. aureus) to quantify intracellular cytotoxic S. aureus phenotypes. Studying a panel of 387 S. aureus bacteraemia isolates, and combined with comparative, statistical, and functional genomics, our platform identified mutations in S. aureus clinical isolates that reduced bacterial cytotoxicity and promoted intracellular persistence. In addition to numerous convergent mutations in the Agr quorum sensing system, our approach detected mutations in other loci that also impacted cytotoxicity and intracellular persistence. We discovered that clinical mutations in ausA, encoding the aureusimine non-ribosomal peptide synthetase, reduced S. aureus cytotoxicity, and increased intracellular persistence. InToxSa is a versatile, high-throughput cell-based phenomics platform and we showcase its utility by identifying clinically relevant S. aureus pathoadaptive mutations that promote intracellular residency.
Collapse
Affiliation(s)
- Abderrahman Hachani
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Stefano G Giulieri
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Romain Guérillot
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Calum J Walsh
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Marion Herisse
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Ye Mon Soe
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Sarah L Baines
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - David R Thomas
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
- Infection and Immunity Program, Department of Microbiology and Biomedicine Discovery Institute, Monash UniversityClaytonAustralia
| | - Shane Doris Cheung
- Biological Optical Microscopy Platform, University of MelbourneMelbourneAustralia
| | - Ashleigh S Hayes
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Ellie Cho
- Biological Optical Microscopy Platform, University of MelbourneMelbourneAustralia
| | - Hayley J Newton
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
- Infection and Immunity Program, Department of Microbiology and Biomedicine Discovery Institute, Monash UniversityClaytonAustralia
| | - Sacha Pidot
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Ruth C Massey
- School of Microbiology, University College CorkCorkIreland
- School of Medicine, University College CorkCorkIreland
- APC Microbiome Ireland, University College CorkCorkIreland
- School of Cellular and Molecular Medicine, University of BristolBristolUnited Kingdom
| | - Benjamin P Howden
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| | - Timothy P Stinear
- Department of Microbiology and Immunology, Doherty Institute, University of MelbourneMelbourneAustralia
| |
Collapse
|
22
|
Cecotto L, Stapels DAC, van Kessel KPM, Croes M, Lourens Z, Vogely HC, van der Wal BCH, van Strijp JAG, Weinans H, Amin Yavari S. Evaluation of silver bio-functionality in a multicellular in vitro model: towards reduced animal usage in implant-associated infection research. Front Cell Infect Microbiol 2023; 13:1186936. [PMID: 37342248 PMCID: PMC10277478 DOI: 10.3389/fcimb.2023.1186936] [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: 04/11/2023] [Accepted: 05/16/2023] [Indexed: 06/22/2023] Open
Abstract
Background Despite the extensive use of silver ions or nanoparticles in research related to preventing implant-associated infections (IAI), their use in clinical practice has been debated. This is because the strong antibacterial properties of silver are counterbalanced by adverse effects on host cells. One of the reasons for this may be the lack of comprehensive in vitro models that are capable of analyzing host-bacteria and host-host interactions. Methods and results In this study, we tested silver efficacy through multicellular in vitro models involving macrophages (immune system), mesenchymal stem cells (MSCs, bone cells), and S. aureus (pathogen). Our model showed to be capable of identifying each element of culture as well as tracking the intracellular survival of bacteria. Furthermore, the model enabled to find a therapeutic window for silver ions (AgNO3) and silver nanoparticles (AgNPs) where the viability of host cells was not compromised, and the antibacterial properties of silver were maintained. While AgNO3 between 0.00017 and 0.017 µg/mL retained antibacterial properties, host cell viability was not affected. The multicellular model, however, demonstrated that those concentrations had no effect on the survival of S. aureus, inside or outside host cells. Similarly, treatment with 20 nm AgNPs did not influence the phagocytic and killing capacity of macrophages or prevent S. aureus from invading MSCs. Moreover, exposure to 100 nm AgNPs elicited an inflammatory response by host cells as detected by the increased production of TNF-α and IL-6. This was visible only when macrophages and MSCs were cultured together. Conclusions Multicellular in vitro models such as the one used here that simulate complex in vivo scenarios can be used to screen other therapeutic compounds or antibacterial biomaterials without the need to use animals.
Collapse
Affiliation(s)
- Leonardo Cecotto
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Daphne A. C. Stapels
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
- Infection Biology Group, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, Netherlands
| | - Kok P. M. van Kessel
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Michiel Croes
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Zeldali Lourens
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - H. Charles Vogely
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Jos A. G. van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Harrie Weinans
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, Netherlands
- Department of Biomechanical Engineering, Delft University of Technology, Delft, Netherlands
| | - Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, Netherlands
- Regenerative Medicine Centre Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
23
|
Guan SW, Lin Q, Yu HB. Intratumour microbiome of pancreatic cancer. World J Gastrointest Oncol 2023; 15:713-730. [PMID: 37275446 PMCID: PMC10237023 DOI: 10.4251/wjgo.v15.i5.713] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/26/2023] [Accepted: 04/04/2023] [Indexed: 05/12/2023] Open
Abstract
Pancreatic cancer is a high mortality malignancy with almost equal mortality and morbidity rates. Both normal and tumour tissues of the pancreas were previously considered sterile. In recent years, with the development of technologies for high-throughput sequencing, a variety of studies have revealed that pancreatic cancer tissues contain small amounts of bacteria and fungi. The intratumour microbiome is being revealed as an influential contributor to carcinogenesis. The intratumour microbiome has been identified as a crucial factor for pancreatic cancer progression, diagnosis, and treatment, chemotherapy resistance, and immune response. A better understanding of the biology of the intratumour microbiome of pancreatic cancer contributes to the establishment of better early cancer screening and treatment strategies. This review focuses on the possible origins of the intratumour microbiome in pancreatic cancer, the intratumour localization, the interaction with the tumour microenvironment, and strategies for improving the outcome of pancreatic cancer treatment. Thus, this review offers new perspectives for improving the prognosis of pancreatic cancer.
Collapse
Affiliation(s)
- Shi-Wei Guan
- Department of Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Quan Lin
- Department of Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Hai-Bo Yu
- Department of Surgery, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| |
Collapse
|
24
|
Maurer M, Klassert TE, Löffler B, Slevogt H, Tuchscherr L. Extraction of High-Quality RNA from S. aureus Internalized by Endothelial Cells. Microorganisms 2023; 11:microorganisms11041020. [PMID: 37110443 PMCID: PMC10143013 DOI: 10.3390/microorganisms11041020] [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: 03/12/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Staphylococcus aureus evades antibiotic therapy and antimicrobial defenses by entering human host cells. Bacterial transcriptomic analysis represents an invaluable tool to unravel the complex interplay between host and pathogen. Therefore, the extraction of high-quality RNA from intracellular S. aureus lays the foundation to acquire meaningful gene expression data. In this study, we present a novel and straightforward strategy to isolate RNA from internalized S. aureus after 90 min, 24 h, and 48 h postinfection. Real-time PCR data were obtained for the target genes agrA and fnba, which play major roles during infection. The commonly used reference genes gyrB, aroE, tmRNA, gmk, and hu were analyzed under different conditions: bacteria from culture (condition I), intracellular bacteria (condition II), and across both conditions I and II. The most stable reference genes were used for the normalization of agrA and fnbA. Delta Cq (quantification cycle) values had a relatively low variability and thus demonstrated the high quality of the extracted RNA from intracellular S. aureus during the early phase of infection. The established protocol allows the extraction and purification of intracellular staphylococcal RNA while minimizing the amount of host RNA in the sample. This approach can leverage reproducible gene expression data to study host-pathogen interactions.
Collapse
Affiliation(s)
- Michelle Maurer
- Institute for Medical Microbiology, Jena University Hospital, 07747 Jena, Germany
| | - Tilman E Klassert
- Department of Respiratory Medicine, Medizinische Hochschule Hannover, 30625 Hannover, Germany
- Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Bettina Löffler
- Institute for Medical Microbiology, Jena University Hospital, 07747 Jena, Germany
| | - Hortense Slevogt
- Department of Respiratory Medicine, Medizinische Hochschule Hannover, 30625 Hannover, Germany
- Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Lorena Tuchscherr
- Institute for Medical Microbiology, Jena University Hospital, 07747 Jena, Germany
| |
Collapse
|
25
|
Rigaill J, Gavid M, Fayolle M, Morgene MF, Lelonge Y, Grattard F, Pozzetto B, Crépin A, Prades JM, Laurent F, Botelho-Nevers E, Berthelot P, Verhoeven PO. Staphylococcus aureus nasal colonization level and intracellular reservoir: a prospective cohort study. Eur J Clin Microbiol Infect Dis 2023; 42:621-629. [PMID: 36964269 DOI: 10.1007/s10096-023-04591-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 03/15/2023] [Indexed: 03/26/2023]
Abstract
Staphylococcus aureus is a major pathogen in humans. The nasal vestibule is considered as the main reservoir of S. aureus. However, even though the nasal cavity may also be colonized by S. aureus, the relationships between the two sites are still unclear. We conducted a prospective study in humans to assess the S. aureus colonization profiles in the vestibule and nasal cavity, and to investigate the presence of intracellular S. aureus in the two sites. Patients undergoing ear, nose, and throat surgery were swabbed during endoscopy to determine S. aureus nasal load, genotype, and presence of intracellular S. aureus. Among per-operative samples from 90 patients, the prevalence of S. aureus carriage was 32.2% and 33.3% in the vestibule and the nasal cavity, respectively. The mean S. aureus load was 4.10 and 4.25 log10 CFU/swab for the nasal vestibule and nasal cavity, respectively (P > 0.05). Genotyping of S. aureus revealed that all nasal strains isolated from a given individual belong to the same clonal complex and spa-type. An intracellular carriage was observed in 5.6% of the patients, all of whom exhibited a S. aureus vestibule load higher than 3 log10 CFU/swab. An intracellular niche was observed in the vestibule as well as in the nasal cavity. In conclusion, the nasal cavity was also found to be a major site of S. aureus carriage in humans and should draw attention when studying host-pathogen interactions related to the risk of infection associated with colonization.
Collapse
Affiliation(s)
- Josselin Rigaill
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Université de Lyon, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, University Jean Monnet, St-Etienne, France
- Department of Infectious Agents and Hygiene, University Hospital of St-Etienne, St-Etienne, France
| | - Marie Gavid
- Department of Ear Nose Throat Surgery, University Hospital of St-Etienne, St-Etienne, France
| | - Martin Fayolle
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Université de Lyon, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, University Jean Monnet, St-Etienne, France
- Department of Infectious Agents and Hygiene, University Hospital of St-Etienne, St-Etienne, France
| | - Mohamed Fedy Morgene
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Université de Lyon, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, University Jean Monnet, St-Etienne, France
| | - Yann Lelonge
- Department of Ear Nose Throat Surgery, University Hospital of St-Etienne, St-Etienne, France
| | - Florence Grattard
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Université de Lyon, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, University Jean Monnet, St-Etienne, France
- Department of Infectious Agents and Hygiene, University Hospital of St-Etienne, St-Etienne, France
| | - Bruno Pozzetto
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Université de Lyon, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, University Jean Monnet, St-Etienne, France
- Department of Infectious Agents and Hygiene, University Hospital of St-Etienne, St-Etienne, France
| | - Adeline Crépin
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Université de Lyon, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, University Jean Monnet, St-Etienne, France
| | - Jean-Michel Prades
- Department of Ear Nose Throat Surgery, University Hospital of St-Etienne, St-Etienne, France
| | - Frédéric Laurent
- CIRI - Centre International de Recherche en Infectiologie, Staphylococcal Pathogenesis team, Université de Lyon, Inserm, U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Department of Bacteriology, Institut des Agents Infectieux, Hospices Civils de Lyon, Lyon, France
| | - Elisabeth Botelho-Nevers
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Université de Lyon, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, University Jean Monnet, St-Etienne, France
- Department of Infectious Diseases, University Hospital of St-Etienne, St-Etienne, France
| | - Philippe Berthelot
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Université de Lyon, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Faculty of Medicine, University Jean Monnet, St-Etienne, France
- Department of Infectious Diseases, University Hospital of St-Etienne, St-Etienne, France
| | - Paul O Verhoeven
- CIRI - Centre International de Recherche en Infectiologie, GIMAP team, Université de Lyon, Inserm, U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.
- Faculty of Medicine, University Jean Monnet, St-Etienne, France.
- Department of Infectious Agents and Hygiene, University Hospital of St-Etienne, St-Etienne, France.
| |
Collapse
|
26
|
vom Werth KL, Kemper B, Kampmeier S, Mellmann A. Application of Digital Holographic Microscopy to Analyze Changes in T-Cell Morphology in Response to Bacterial Challenge. Cells 2023; 12:cells12050762. [PMID: 36899897 PMCID: PMC10000559 DOI: 10.3390/cells12050762] [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: 01/25/2023] [Revised: 02/16/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
Quantitative phase imaging (QPI) is a non-invasive, label-free technique used to detect aberrant cell morphologies caused by disease, thus providing a useful diagnostic approach. Here, we evaluated the potential of QPI to differentiate specific morphological changes in human primary T-cells exposed to various bacterial species and strains. Cells were challenged with sterile bacterial determinants, i.e., membrane vesicles or culture supernatants, derived from different Gram-positive and Gram-negative bacteria. Timelapse QPI by digital holographic microscopy (DHM) was applied to capture changes in T-cell morphology over time. After numerical reconstruction and image segmentation, we calculated single cell area, circularity and mean phase contrast. Upon bacterial challenge, T-cells underwent rapid morphological changes such as cell shrinkage, alterations of mean phase contrast and loss of cell integrity. Time course and intensity of this response varied between both different species and strains. The strongest effect was observed for treatment with S. aureus-derived culture supernatants that led to complete lysis of the cells. Furthermore, cell shrinkage and loss of circular shape was stronger in Gram-negative than in Gram-positive bacteria. Additionally, T-cell response to bacterial virulence factors was concentration-dependent, as decreases in cellular area and circularity were enhanced with increasing concentrations of bacterial determinants. Our findings clearly indicate that T-cell response to bacterial stress depends on the causative pathogen, and specific morphological alterations can be detected using DHM.
Collapse
Affiliation(s)
| | - Björn Kemper
- Biomedical Technology Center of the Medical Faculty, University of Münster, 48149 Münster, Germany
| | - Stefanie Kampmeier
- Institute of Hygiene, University Hospital Münster, 48149 Münster, Germany
| | - Alexander Mellmann
- Institute of Hygiene, University Hospital Münster, 48149 Münster, Germany
- Correspondence: ; Tel.: +49-251-83-55361
| |
Collapse
|
27
|
Otto M. Critical Assessment of the Prospects of Quorum-Quenching Therapy for Staphylococcus aureus Infection. Int J Mol Sci 2023; 24:ijms24044025. [PMID: 36835436 PMCID: PMC9958572 DOI: 10.3390/ijms24044025] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/04/2023] [Accepted: 02/08/2023] [Indexed: 02/19/2023] Open
Abstract
Staphylococcus aureus is an important pathogen that causes a high number of infections and is one of the leading causes of death in hospitalized patients. Widespread antibiotic resistance such as in methicillin-resistant S. aureus (MRSA) has prompted research into potential anti-virulence-targeted approaches. Targeting the S. aureus accessory gene regulator (Agr) quorum-sensing system, a master regulator of virulence, is the most frequently proposed anti-virulence strategy for S. aureus. While much effort has been put into the discovery and screening for Agr inhibitory compounds, in vivo analysis of their efficacy in animal infection models is still rare and reveals various shortcomings and problems. These include (i) an almost exclusive focus on topical skin infection models, (ii) technical problems that leave doubt as to whether observed in vivo effects are due to quorum-quenching, and (iii) the discovery of counterproductive biofilm-increasing effects. Furthermore, potentially because of the latter, invasive S. aureus infection is associated with Agr dysfunctionality. Altogether, the potential of Agr inhibitory drugs is nowadays seen with low enthusiasm given the failure to provide sufficient in vivo evidence for their potential after more than two decades since the initiation of such efforts. However, current Agr inhibition-based probiotic approaches may lead to a new application of Agr inhibition strategies in preventing S. aureus infections by targeting colonization or for otherwise difficult-to-treat skin infections such as atopic dermatitis.
Collapse
Affiliation(s)
- Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814, USA
| |
Collapse
|
28
|
MRSA Isolates from Patients with Persistent Bacteremia Generate Nonstable Small Colony Variants In Vitro within Macrophages and Endothelial Cells during Prolonged Vancomycin Exposure. Infect Immun 2023; 91:e0042322. [PMID: 36602380 PMCID: PMC9872686 DOI: 10.1128/iai.00423-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Staphylococcus aureus (especially methicillin-resistant S. aureus [MRSA]) is frequently associated with persistent bacteremia (PB) during vancomycin therapy despite consistent susceptibility in vitro. Strategic comparisons of PB strains versus those from vancomycin-resolving bacteremia (RB) would yield important mechanistic insights into PB outcomes. Clinical PB versus RB isolates were assessed in vitro for intracellular replication and small colony variant (SCV) formation within macrophages and endothelial cells (ECs) in the presence or absence of exogenous vancomycin. In both macrophages and ECs, PB and RB isolates replicated within lysosome-associated membrane protein-1 (LAMP-1)-positive compartments. PB isolates formed nonstable small colony variants (nsSCVs) in vancomycin-exposed host cells at a significantly higher frequency than matched RB isolates (in granulocyte-macrophage colony-stimulating factor [GM-CSF], human macrophages PB versus RB, P < 0.0001 at 48 h; in ECs, PB versus RB, P < 0.0001 at 24 h). This phenotype could represent one potential basis for the unique ability of PB isolates to adaptively resist vancomycin therapy and cause PB in humans. Elucidating the molecular mechanism(s) by which PB strains form nsSCVs could facilitate the discovery of novel treatment strategies to mitigate PB due to MRSA.
Collapse
|
29
|
Sun L, Zhang H, Zhang H, Lou X, Wang Z, Wu Y, Yang X, Chen D, Guo B, Zhang A, Qian F. Staphylococcal virulence factor HlgB targets the endoplasmic-reticulum-resident E3 ubiquitin ligase AMFR to promote pneumonia. Nat Microbiol 2023; 8:107-120. [PMID: 36593296 DOI: 10.1038/s41564-022-01278-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 10/21/2022] [Indexed: 01/03/2023]
Abstract
Staphylococcus aureus invades cells and persists intracellularly, causing persistent inflammation that is notoriously difficult to treat. Here we investigated host-pathogen interactions underlying intracellular S. aureus infection in macrophages and discovered that the endoplasmic reticulum (ER) is an important cellular compartment for intracellular S. aureus infection. Using CRISPR-Cas9 guide RNA library screening, we determined that the autocrine motility factor receptor (AMFR), an ER-resident E3 ubiquitin ligase, played an essential role in mediating intracellular S. aureus-induced inflammation. AMFR directly interacted with TAK1-binding protein 3 (TAB3) in the ER, inducing K27-linked polyubiquitination of TAB3 on lysine 649 and promoting TAK1 activation. Moreover, the virulence factor γ-haemolysin B (HIgB) of S. aureus bound to the AMFR and regulated TAB3. Our findings highlight an unknown role of AMFR in intracellular S. aureus infection-induced pneumonia and suggest that pharmacological interruption of AMFR-mediated TAB3 signalling cascades and HIgB targeting may prevent invasive staphylococci-mediated pneumonia.
Collapse
Affiliation(s)
- Lei Sun
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, Research Center for Small Molecule Immunological Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China.
| | - Haibo Zhang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, Research Center for Small Molecule Immunological Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Huihui Zhang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, Research Center for Small Molecule Immunological Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Xinyi Lou
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, Research Center for Small Molecule Immunological Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiming Wang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, Research Center for Small Molecule Immunological Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yaxian Wu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xinyi Yang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, Research Center for Small Molecule Immunological Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Daijie Chen
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, Research Center for Small Molecule Immunological Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Beining Guo
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China
| | - Ao Zhang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, Research Center for Small Molecule Immunological Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Qian
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, Research Center for Small Molecule Immunological Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
30
|
Jorde I, Schreiber J, Stegemann-Koniszewski S. The Role of Staphylococcus aureus and Its Toxins in the Pathogenesis of Allergic Asthma. Int J Mol Sci 2022; 24:ijms24010654. [PMID: 36614093 PMCID: PMC9820472 DOI: 10.3390/ijms24010654] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 01/01/2023] Open
Abstract
Bronchial asthma is one of the most common chronic diseases worldwide and affects more than 300 million patients. Allergic asthma affects the majority of asthmatic children as well as approximately 50% of adult asthmatics. It is characterized by a Th2-mediated immune response against aeroallergens. Many aspects of the overall pathophysiology are known, while the underlying mechanisms and predisposing factors remain largely elusive today. Over the last decade, respiratory colonization with Staphylococcus aureus (S. aureus), a Gram-positive facultative bacterial pathogen, came into focus as a risk factor for the development of atopic respiratory diseases. More than 30% of the world’s population is constantly colonized with S. aureus in their nasopharynx. This colonization is mostly asymptomatic, but in immunocompromised patients, it can lead to serious complications including pneumonia, sepsis, or even death. S. aureus is known for its ability to produce a wide range of proteins including toxins, serine-protease-like proteins, and protein A. In this review, we provide an overview of the current knowledge about the pathophysiology of allergic asthma and to what extent it can be affected by different toxins produced by S. aureus. Intensifying this knowledge might lead to new preventive strategies for atopic respiratory diseases.
Collapse
|
31
|
Raineri EJM, Maaß S, Wang M, Brushett S, Palma Medina LM, Sampol Escandell N, Altulea D, Raangs E, de Jong A, Vera Murguia E, Feil EJ, Friedrich AW, Buist G, Becher D, García-Cobos S, Couto N, van Dijl JM. Staphylococcus aureus populations from the gut and the blood are not distinguished by virulence traits-a critical role of host barrier integrity. MICROBIOME 2022; 10:239. [PMID: 36567349 PMCID: PMC9791742 DOI: 10.1186/s40168-022-01419-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/09/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND The opportunistic pathogen Staphylococcus aureus is an asymptomatically carried member of the microbiome of about one third of the human population at any given point in time. Body sites known to harbor S. aureus are the skin, nasopharynx, and gut. In particular, the mechanisms allowing S. aureus to pass the gut epithelial barrier and to invade the bloodstream were so far poorly understood. Therefore, the objective of our present study was to investigate the extent to which genetic differences between enteric S. aureus isolates and isolates that caused serious bloodstream infections contribute to the likelihood of invasive disease. RESULTS Here, we present genome-wide association studies (GWAS) that compare the genome sequences of 69 S. aureus isolates from enteric carriage by healthy volunteers and 95 isolates from bloodstream infections. We complement our GWAS results with a detailed characterization of the cellular and extracellular proteomes of the representative gut and bloodstream isolates, and by assaying the virulence of these isolates with infection models based on human gut epithelial cells, human blood cells, and a small animal infection model. Intriguingly, our results show that enteric and bloodstream isolates with the same sequence type (ST1 or ST5) are very similar to each other at the genomic and proteomic levels. Nonetheless, bloodstream isolates are not necessarily associated with an invasive profile. Furthermore, we show that the main decisive factor preventing infection of gut epithelial cells in vitro is the presence of a tight barrier. CONCLUSIONS Our data show that virulence is a highly variable trait, even within a single clone. Importantly, however, there is no evidence that blood stream isolates possess a higher virulence potential than those from the enteric carriage. In fact, some gut isolates from healthy carriers were more virulent than bloodstream isolates. Based on our present observations, we propose that the integrity of the gut epithelial layer, rather than the pathogenic potential of the investigated enteric S. aureus isolates, determines whether staphylococci from the gut microbiome will become invasive pathogens. Video Abstract.
Collapse
Affiliation(s)
- Elisa J. M. Raineri
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sandra Maaß
- Department of Microbial Proteomics, Institute of Microbiology, University of Greifswald, Greifswald, Germany
| | - Min Wang
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Siobhan Brushett
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Laura M. Palma Medina
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Present address: Department of Medicine Huddinge, Present Address: Center for Infectious Medicine, Karolinska Institute, Huddinge, Sweden
| | - Neus Sampol Escandell
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dania Altulea
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Present address: Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Erwin Raangs
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anne de Jong
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen, The Netherlands
| | - Elias Vera Murguia
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Edward J. Feil
- Department of Biology and Biochemistry, The Milner Centre for Evolution, University of Bath, Bath, UK
| | - Alex W. Friedrich
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Girbe Buist
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dörte Becher
- Department of Microbial Proteomics, Institute of Microbiology, University of Greifswald, Greifswald, Germany
| | - Silvia García-Cobos
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Present address: Reference and Research Laboratory On Antimicrobial Resistance and Healthcare Associated Infections, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Natacha Couto
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Biology and Biochemistry, The Milner Centre for Evolution, University of Bath, Bath, UK
| | - Jan Maarten van Dijl
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
32
|
Johnson MB, Furr KH, Suptela SR, Leach W, Marriott I. Induction of protective interferon-β responses in murine osteoblasts following Staphylococcus aureus infection. Front Microbiol 2022; 13:1066237. [PMID: 36532419 PMCID: PMC9757064 DOI: 10.3389/fmicb.2022.1066237] [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: 10/10/2022] [Accepted: 11/16/2022] [Indexed: 12/05/2022] Open
Abstract
Introduction The refractory and recurrent nature of chronic staphylococcal osteomyelitis may be due, at least in part, to the ability of Staphylococcus aureus to invade and persist within bone-forming osteoblasts. However, osteoblasts are now recognized to respond to S. aureus infection and produce numerous immune mediators and bone regulatory factors that can shape the host response. Type I interferons (IFNs) are best known for their antiviral effects, but it is becoming apparent that they impact host susceptibility to a wide range of pathogens including S. aureus. Methods Here, we have assessed the local expression of IFN-β by specific capture ELISA in an established in vivo mouse model of staphylococcal osteomyelitis. RNA Tag-Seq analysis, specific capture ELISAs, and/or immunoblot analyses, were then used to assess the expression of type I IFNs and select IFN stimulated genes (ISGs) in S. aureus infected primary murine osteoblasts. The effect of IFN-β on intracellular S. aureus burden was assessed in vitro following recombinant cytokine treatment by serial colony counts of liberated bacteria. Results We report the presence of markedly elevated IFN-β levels in infected bone tissue in a mouse model of staphylococcal osteomyelitis. RNA Tag-Seq analysis of S. aureus infected osteoblasts showed enrichment of genes associated with type I IFN signaling and ISGs, and elevated expression of mRNA encoding IFN-β and ISG products. IFN-β production was confirmed with the demonstration that S. aureus induces its rapid and robust release by osteoblasts in a dose-dependent manner. Furthermore, we showed increased protein expression of the ISG products IFIT1 and IFIT3 by infected osteoblasts and demonstrate that this occurs secondary to the release of IFN-β by these cells. Finally, we have determined that exposure of S. aureus-infected osteoblasts to IFN-β markedly reduces the number of viable bacteria harbored by these cells. Discussion Together, these findings indicate an ability of osteoblasts to respond to bacteria by producing IFN-β that can act in an autocrine and/or paracrine manner to elicit ISG expression and mitigate S. aureus infection.
Collapse
Affiliation(s)
- M. Brittany Johnson
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, United States
| | - Kelli H. Furr
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, United States
| | - Samantha R. Suptela
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, United States
| | - Whitney Leach
- Department of Molecular Biology, Stowers Institute for Medical Research, Kansas City, MO, United States
| | - Ian Marriott
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, United States
| |
Collapse
|
33
|
Billings C, Rifkin R, Abouelkhair M, Jones RD, Bow A, Kolape J, Rajeev S, Kania S, Anderson DE. In vitro and in vivo assessment of caprine origin Staphylococcus aureus ST398 strain UTCVM1 as an osteomyelitis pathogen. Front Cell Infect Microbiol 2022; 12:1015655. [PMID: 36726643 PMCID: PMC9885270 DOI: 10.3389/fcimb.2022.1015655] [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: 08/09/2022] [Accepted: 10/17/2022] [Indexed: 11/25/2022] Open
Abstract
Staphylococcus aureus (SA) is a significant and well-recognized causative organism of bacterial osteomyelitis. Osteomyelitis is an inflammatory bone disease characterized by progressive bone destruction and loss. This disease causes significant morbidity and mortality to the patient and poses therapeutic challenges for clinicians. To improve the efficacy of therapeutic strategies to combat bacterial osteomyelitis, there is a need to define the molecular epidemiology of bacterial organisms more clearly and further the understanding of the pathogenesis of SA osteomyelitis. We conducted in vitro characterization of the pathogenic capabilities of an isolate of SA ST398 derived from a clinical case of osteomyelitis in a goat. We also report a rodent mandibular defect model to determine the ability of ST398 to cause reproducible osteomyelitis. Our results indicate that ST398 can invade and distort pre-osteoblastic cells in culture, induce significant inflammation and alter expression of osteoregulatory cytokines. We also demonstrate the ability of ST398 to induce osteomyelitis in a rat mandibular model. When compiled, these data support ST398 as a competent osteomyelitis pathogen.
Collapse
Affiliation(s)
- Caroline Billings
- Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, United States,*Correspondence: Caroline Billings,
| | - Rebecca Rifkin
- Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, United States
| | - Mohamed Abouelkhair
- Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, United States
| | - Rebekah Duckett Jones
- Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, United States
| | - Austin Bow
- Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, United States
| | - Jaydeep Kolape
- Advanced Microscopy and Imaging Center, University of Tennessee, Knoxville, TN, United States
| | - Sreekumari Rajeev
- Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, United States
| | - Stephen Kania
- Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, United States
| | - David E. Anderson
- Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, United States
| |
Collapse
|
34
|
Suthi S, Gopi D, Chaudhary A, Sarma PVGK. The Therapeutic Potential of 4-Methoxy-1-methyl-2-oxopyridine-3-carbamide (MMOXC) Derived from Ricinine on Macrophage Cell Lines Infected with Methicillin-Resistant Strains of Staphylococcus aureus. Appl Biochem Biotechnol 2022; 195:2843-2862. [PMID: 36418711 DOI: 10.1007/s12010-022-04269-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2022] [Indexed: 11/25/2022]
Abstract
The incidences of methicillin-resistant strains of Staphylococcus aureus (MRSA) and their survival inside the macrophages are the major attributes of the relapsed infections after antimicrobial therapy, and it is a global problem. In this context, we have previously demonstrated 4-methoxy-1-methyl-2-oxopyridine-3-carbamide (MMOXC), a Ricinine derivative exhibiting anti-S. aureus and anti-biofilm characteristics by competitively inhibiting uridine monophosphate kinase (UMPK), UDP-N-acetyl muramyl pentapeptide ligase (Mur-F), and peptidyl deformylase, (PDF). In the present study, the stability of this competitive inhibitor MMOXC was evaluated by showing its ability to remain bound to the active sites of UMPK, Mur-F, and PDF even after increasing the incubation time, temperature, pH, and substrate concentration. On growing MRSA in fewer concentrations of MMOXC, these strains could not attain resistance to MMOXC and at the same time distinct reductions in the expression of UMPK, Mur-F, and PDF genes were noted. In vitro, infective models were generated by infecting MRSA to RAW 264.7 and human monocyte-derived macrophage (hMDM) cell lines. In these infected cell lines, in spite of increased nitric oxide synthase (NOS), NADPH-P450 reductase, superoxide dismutase, catalase, and peroxidase activities, the MRSA survived. At 640 µM/ml, the concentration of MMOXC penetrated into these infected cells and obliterated MRSA. While treating uninfected macrophage cell lines with MMOXC, no appreciable effect was observed indicating that MMOXC is the most suitable drug for the treatment of infections caused by MRSA.
Collapse
Affiliation(s)
- Subbarayudu Suthi
- Microbial Genetics Laboratory, Department of Biotechnology, Sri Venkateswara Institute of Medical Sciences and University, Alipiri Road, Andhra Pradesh, 517501, Tirupati, India
| | - Deepika Gopi
- Microbial Genetics Laboratory, Department of Biotechnology, Sri Venkateswara Institute of Medical Sciences and University, Alipiri Road, Andhra Pradesh, 517501, Tirupati, India
| | - Abhijit Chaudhary
- Department of Microbiology, Sri Venkateswara Institute of Medical Sciences, Tirupati, 517501, Andhra Pradesh, India
| | - Potukuchi Venkata Gurunadha Krishna Sarma
- Microbial Genetics Laboratory, Department of Biotechnology, Sri Venkateswara Institute of Medical Sciences and University, Alipiri Road, Andhra Pradesh, 517501, Tirupati, India.
| |
Collapse
|
35
|
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: 8] [Impact Index Per Article: 2.7] [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
|
36
|
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
|
37
|
Liu K, Zhou X, Fang L, Dong J, Cui L, Li J, Meng X, Zhu G, Li J, Wang H. PINK1/parkin-mediated mitophagy alleviates Staphylococcus aureus-induced NLRP3 inflammasome and NF-κB pathway activation in bovine mammary epithelial cells. Int Immunopharmacol 2022; 112:109200. [PMID: 36063687 DOI: 10.1016/j.intimp.2022.109200] [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] [Received: 07/11/2022] [Revised: 08/18/2022] [Accepted: 08/23/2022] [Indexed: 11/28/2022]
Abstract
Staphylococcus aureus (S. aureus) is known to induce chronic and persistent bovine mammary infection, which affects milk quality and leads to premature culling. The ability of S. aureus to invade mammalian cells protects it from clearance by the immune system. Mitophagy is important in cell homeostasis, and can be utilized by pathogens for immune escape. However, mitophagy's role in S. aureus-associated bovine mastitis remains unclear. Here, S. aureus infection induced mitophagy and enhanced mitochondrial translocation of parkin in MAC-T cells. After mitophagy inhibition by Mdivi-1 treatment or PTEN-induced putative kinase 1 (PINK1) silencing in MAC-T cells infected with S. aureus, NOD-like receptor protein 3 (NLRP3) inflammasome activation and p65 and IκBα phosphorylation were increased. Meanwhile, PINK1 overexpression had the opposite effects. In addition, NLRP3 inflammasome overactivation and enhanced p65 and IκBα phosphorylation caused by PINK1 silencing were reversed by MitoTEMPO. Furthermore, PINK1/parkin-mediated mitophagy promoted S. aureus survival and contributed to persistent S. aureus infection. These findings provide new insights into S. aureus invasion in bovine mastitis.
Collapse
Affiliation(s)
- Kangjun Liu
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Important Animal Infectious Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, Jiangsu 225009, China.
| | - Xi Zhou
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Important Animal Infectious Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, Jiangsu 225009, China.
| | - Li Fang
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Important Animal Infectious Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, Jiangsu 225009, China.
| | - Junsheng Dong
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Important Animal Infectious Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, Jiangsu 225009, China.
| | - Luying Cui
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Important Animal Infectious Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, Jiangsu 225009, China.
| | - Jun Li
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Important Animal Infectious Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, Jiangsu 225009, China.
| | - Xia Meng
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Important Animal Infectious Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, Jiangsu 225009, China.
| | - Guoqiang Zhu
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Important Animal Infectious Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, Jiangsu 225009, China.
| | - Jianji Li
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Important Animal Infectious Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, Jiangsu 225009, China.
| | - Heng Wang
- College of Veterinary Medicine, Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou, Jiangsu 225009, China; Joint International Research Laboratory of Important Animal Infectious Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, Jiangsu 225009, China.
| |
Collapse
|
38
|
Zelmer AR, Nelson R, Richter K, Atkins GJ. Can intracellular Staphylococcus aureus in osteomyelitis be treated using current antibiotics? A systematic review and narrative synthesis. Bone Res 2022; 10:53. [PMID: 35961964 PMCID: PMC9374758 DOI: 10.1038/s41413-022-00227-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/26/2022] [Accepted: 06/15/2022] [Indexed: 11/09/2022] Open
Abstract
Approximately 40% of treatments of chronic and recurrent osteomyelitis fail in part due to bacterial persistence. Staphylococcus aureus, the predominant pathogen in human osteomyelitis, is known to persist by phenotypic adaptation as small-colony variants (SCVs) and by formation of intracellular reservoirs, including those in major bone cell types, reducing susceptibility to antibiotics. Intracellular infections with S. aureus are difficult to treat; however, there are no evidence-based clinical guidelines addressing these infections in osteomyelitis. We conducted a systematic review of the literature to determine the demonstrated efficacy of all antibiotics against intracellular S. aureus relevant to osteomyelitis, including protein biosynthesis inhibitors (lincosamides, streptogramins, macrolides, oxazolidines, tetracyclines, fusidic acid, and aminoglycosides), enzyme inhibitors (fluoroquinolones and ansamycines), and cell wall inhibitors (beta-lactam inhibitors, glycopeptides, fosfomycin, and lipopeptides). The PubMed and Embase databases were screened for articles related to intracellular S. aureus infections that compared the effectiveness of multiple antibiotics or a single antibiotic together with another treatment, which resulted in 34 full-text articles fitting the inclusion criteria. The combined findings of these studies were largely inconclusive, most likely due to the plethora of methodologies utilized. Therefore, the reported findings in the context of the models employed and possible solutions for improved understanding are explored here. While rifampicin, oritavancin, linezolid, moxifloxacin and oxacillin were identified as the most effective potential intracellular treatments, the scientific evidence for these is still relatively weak. We advocate for more standardized research on determining the intracellular effectiveness of antibiotics in S. aureus osteomyelitis to improve treatments and patient outcomes.
Collapse
Affiliation(s)
- Anja R Zelmer
- Centre for Orthopaedic and Trauma Research, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Renjy Nelson
- Department of Infectious Diseases, Central Adelaide Local Health Network, Adelaide, SA, 5000, Australia.,Royal Adelaide Hospital, Adelaide, SA, 5000, Australia
| | - Katharina Richter
- Richter Lab, Department of Surgery, Basil Hetzel Institute for Translational Health Research, University of Adelaide, Adelaide, SA, 5011, Australia
| | - Gerald J Atkins
- Centre for Orthopaedic and Trauma Research, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, 5000, Australia.
| |
Collapse
|
39
|
A Model of Intracellular Persistence of Pseudomonas aeruginosa in Airway Epithelial Cells. Cell Microbiol 2022. [DOI: 10.1155/2022/5431666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Pseudomonas aeruginosa (P.a.) is a major human pathogen capable of causing chronic infections in hosts with weakened barrier functions and host defenses, most notably airway infections commonly observed in individuals with the genetic disorder cystic fibrosis (CF). While mainly described as an extracellular pathogen, previous in vitro studies have described the molecular events leading to P.a. internalization in diverse epithelial cell types. However, the long-term fate of intracellular P.a. remains largely unknown. Here, we developed a model allowing for a better understanding of long-term (up to 120 h) intracellular bacterial survival in the airway epithelial cell line BEAS-2B. Using a tobramycin protection assay, we characterized the internalization, long-term intracellular survival, and cytotoxicity of the lab strain PAO1, as well as clinical CF isolates, and conducted analyses at the single-cell level using confocal microscopy and flow cytometry techniques. We observed that infection at low multiplicity of infection allows for intracellular survival up to 120 h post-infection without causing significant host cytotoxicity. Finally, infection with clinical isolates revealed significant strain-to-strain heterogeneity in intracellular survival, including a high persistence phenotype associated with bacterial replication within host cells. Future studies using this model will further elucidate the host and bacterial mechanisms that promote P. aeruginosa intracellular persistence in airway epithelial cells, a potentially unrecognized bacterial reservoir during chronic infections.
Collapse
|
40
|
Moreno-Mendieta S, Guillén D, Vasquez-Martínez N, Hernández-Pando R, Sánchez S, Rodríguez-Sanoja R. Understanding the Phagocytosis of Particles: the Key for Rational Design of Vaccines and Therapeutics. Pharm Res 2022; 39:1823-1849. [PMID: 35739369 DOI: 10.1007/s11095-022-03301-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 05/23/2022] [Indexed: 12/17/2022]
Abstract
A robust comprehension of phagocytosis is crucial for understanding its importance in innate immunity. A detailed description of the molecular mechanisms that lead to the uptake and clearance of endogenous and exogenous particles has helped elucidate the role of phagocytosis in health and infectious or autoimmune diseases. Furthermore, knowledge about this cellular process is important for the rational design and development of particulate systems for the administration of vaccines or therapeutics. Depending on these specific applications and the required biological responses, particles must be designed to encourage or avoid their phagocytosis and prolong their circulation time. Functionalization with specific polymers or ligands and changes in the size, shape, or surface of particles have important effects on their recognition and internalization by professional and nonprofessional phagocytes and have a major influence on their fate and safety. Here, we review the phagocytosis of particles intended to be used as carrier or delivery systems for vaccines or therapeutics, the cells involved in this process depending on the route of administration, and the strategies employed to obtain the most desirable particles for each application through the manipulation of their physicochemical characteristics. We also offer a view of the challenges and potential opportunities in the field and give some recommendations that we expect will enable the development of improved approaches for the rational design of these systems.
Collapse
Affiliation(s)
- Silvia Moreno-Mendieta
- Consejo Nacional de Ciencia y Tecnología (CONACyT), Ciudad de México, Mexico. .,Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico.
| | - Daniel Guillén
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico
| | - Nathaly Vasquez-Martínez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico.,Doctorado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico
| | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Delegación Tlalpan, Ciudad de México, Mexico
| | - Sergio Sánchez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico
| | - Romina Rodríguez-Sanoja
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico.
| |
Collapse
|
41
|
Li H, Sun P. Insight of Melatonin: The Potential of Melatonin to Treat Bacteria-Induced Mastitis. Antioxidants (Basel) 2022; 11:antiox11061107. [PMID: 35740004 PMCID: PMC9219804 DOI: 10.3390/antiox11061107] [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: 04/08/2022] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 02/01/2023] Open
Abstract
Bovine mastitis is a common inflammatory disease, mainly induced by bacterial pathogens, such as Staphylococcus aureus, Escherichia coli, and Streptococcus agalactiae. Mastitis has negative effects on the production and quality of milk, resulting in huge economic losses. Melatonin, which is synthesized and secreted by the pineal gland and other organs, is ubiquitous throughout nature and has different effects on different tissues. Melatonin is crucial in modulating oxidative stress, immune responses, and cell autophagy and apoptosis, via receptor-mediated or receptor-independent signaling pathways. The potent antioxidative and anti-inflammatory activities of melatonin and its metabolites suggest that melatonin can be used to treat various infections. This article reviews the potential for melatonin to alleviate bovine mastitis through its pleiotropic effect on reducing oxidative stress, inhibiting pro-inflammatory cytokines, and regulating the activation of NF-κB, STATs, and their cascade reactions. Therefore, it is promising that melatonin supplementation may be an alternative to antibiotics for the treatment of bovine mastitis.
Collapse
|
42
|
Making Sense of Quorum Sensing at the Intestinal Mucosal Interface. Cells 2022; 11:cells11111734. [PMID: 35681429 PMCID: PMC9179481 DOI: 10.3390/cells11111734] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/17/2022] [Accepted: 05/18/2022] [Indexed: 12/12/2022] Open
Abstract
The gut microbiome can produce metabolic products that exert diverse activities, including effects on the host. Short chain fatty acids and amino acid derivatives have been the focus of many studies, but given the high microbial density in the gastrointestinal tract, other bacterial products such as those released as part of quorum sensing are likely to play an important role for health and disease. In this review, we provide of an overview on quorum sensing (QS) in the gastrointestinal tract and summarise what is known regarding the role of QS molecules such as auto-inducing peptides (AIP) and acyl-homoserine lactones (AHL) from commensal, probiotic, and pathogenic bacteria in intestinal health and disease. QS regulates the expression of numerous genes including biofilm formation, bacteriocin and toxin secretion, and metabolism. QS has also been shown to play an important role in the bacteria–host interaction. We conclude that the mechanisms of action of QS at the intestinal neuro–immune interface need to be further investigated.
Collapse
|
43
|
Kremlitzka M, Colineau L, Nowacka AA, Mohlin FC, Wozniak K, Blom AM, King BC. Alternative translation and retrotranslocation of cytosolic C3 that detects cytoinvasive bacteria. Cell Mol Life Sci 2022; 79:291. [PMID: 35546365 PMCID: PMC9095555 DOI: 10.1007/s00018-022-04308-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/29/2022] [Accepted: 04/12/2022] [Indexed: 01/05/2023]
Abstract
Complement C3 was originally regarded as a serum effector protein, although recent data has emerged suggesting that intracellular C3 can also regulate basic cellular processes. Despite the growing interest in intracellular C3 functions, the mechanism behind its generation has not been demonstrated. In this study we show that C3 can be expressed from an alternative translational start site, resulting in C3 lacking the signal peptide, which is therefore translated in the cytosol. In contrast to the secreted form, alternatively translated cytosolic C3 is not glycosylated, is present mainly in a reduced state, and is turned over by the ubiquitin–proteasome system. C3 can also be retrotranslocated from the endoplasmic reticulum into the cytosol, structurally resembling secreted C3. Finally, we demonstrate that intracellular cytosolic C3 can opsonize invasive Staphylococcus aureus within epithelial cell, slowing vacuolar escape as well as impacting bacterial survival on subsequent exposure to phagocytes. Our work therefore reveals the existence and origin of intracellular, cytosolic C3, and demonstrates functions for cytosolic C3 in intracellular detection of cytoinvasive pathogens.
Collapse
Affiliation(s)
- Mariann Kremlitzka
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden.,Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | - Lucie Colineau
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Alicja A Nowacka
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Frida C Mohlin
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Katarzyna Wozniak
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Anna M Blom
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden.
| | - Ben C King
- Division of Medical Protein Chemistry, Department of Translational Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
44
|
Fu A, Yao B, Dong T, Chen Y, Yao J, Liu Y, Li H, Bai H, Liu X, Zhang Y, Wang C, Guo Y, Li N, Cai S. Tumor-resident intracellular microbiota promotes metastatic colonization in breast cancer. Cell 2022; 185:1356-1372.e26. [PMID: 35395179 DOI: 10.1016/j.cell.2022.02.027] [Citation(s) in RCA: 397] [Impact Index Per Article: 132.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/10/2022] [Accepted: 02/24/2022] [Indexed: 12/12/2022]
Abstract
Tumor-resident intracellular microbiota is an emerging tumor component that has been documented for a variety of cancer types with unclear biological functions. Here, we explored the functional significance of these intratumor bacteria, primarily using a murine spontaneous breast-tumor model MMTV-PyMT. We found that depletion of intratumor bacteria significantly reduced lung metastasis without affecting primary tumor growth. During metastatic colonization, intratumor bacteria carried by circulating tumor cells promoted host-cell survival by enhancing resistance to fluid shear stress by reorganizing actin cytoskeleton. We further showed that intratumor administration of selected bacteria strains isolated from tumor-resident microbiota promoted metastasis in two murine tumor models with significantly different levels of metastasis potential. Our findings suggest that tumor-resident microbiota, albeit at low biomass, play an important role in promoting cancer metastasis, intervention of which might therefore be worth exploring for advancing oncology care.
Collapse
Affiliation(s)
- Aikun Fu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China
| | - Bingqing Yao
- School of Life Sciences, Fudan University, Shanghai, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Tingting Dong
- School of Life Sciences, Fudan University, Shanghai, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Yongyi Chen
- Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, 310000, China
| | - Jia Yao
- Department of Breast Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Yu Liu
- Department of Breast Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Hang Li
- Westlake University High-Performance Computing Center, Westlake University, Hangzhou, Zhejiang, China
| | - Huiru Bai
- School of Life Sciences, Fudan University, Shanghai, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Xiaoqin Liu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; School of Life Sciences, Fudan University, Shanghai, China
| | - Yue Zhang
- School of Life Sciences, Fudan University, Shanghai, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Chunhui Wang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Disease Modeling lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Yajing Guo
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Nan Li
- Westlake University High-Performance Computing Center, Westlake University, Hangzhou, Zhejiang, China
| | - Shang Cai
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Disease Modeling lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
45
|
Akoua-Koffi C, Kacou N’Douba A, Djaman JA, Herrmann M, Schaumburg F, Niemann S. Staphylococcus schweitzeri—An Emerging One Health Pathogen? Microorganisms 2022; 10:microorganisms10040770. [PMID: 35456820 PMCID: PMC9026344 DOI: 10.3390/microorganisms10040770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 03/28/2022] [Accepted: 03/31/2022] [Indexed: 11/22/2022] Open
Abstract
The Staphylococcus aureus-related complex is formed by the Staphylococcus aureus, Staphylococcus schweitzeri, Staphylococcus argenteus, Staphylococcus roterodami and Staphylococcus singaporensis. Within this complex, S. schweitzeri is the only species mainly found in African wildlife, but it is rarely detected as a colonizer in humans or as a contaminant of fomites. The few detections in humans are most likely spillover events after contact with wildlife. However, since S. schweitzeri can be misidentified as S. aureus using culture-based routine techniques, it is likely that S. schweitzeri is under-reported in humans. The low number of isolates in humans, though, is consistent with the fact that the pathogen has typical animal adaptation characteristics (e.g., growth kinetics, lack of immune evasion cluster and antimicrobial resistance); however, evidence from selected in vitro assays (e.g., host cell invasion, cell activation, cytotoxicity) indicate that S. schweitzeri might be as virulent as S. aureus. In this case, contact with animals colonized with S. schweitzeri could constitute a risk for zoonotic infections. With respect to antimicrobial resistance, all described isolates were found to be susceptible to all antibiotics tested, and so far no data on the development of spontaneous resistance or the acquisition of resistance genes such the mecA/mecC cassette are available. In summary, general knowledge about this pathogen, specifically on the potential threat it may incur to human and animal health, is still very poor. In this review article, we compile the present state of scientific research, and identify the knowledge gaps that need to be filled in order to reliably assess S. schweitzeri as an organism with global One Health implications.
Collapse
Affiliation(s)
- Chantal Akoua-Koffi
- Centre Hospitalier Universitaire de Bouaké, Bouaké P.O. Box BP 1174, Côte d’Ivoire;
- Department of Biology, Université Alassane Ouattara de Bouaké, Bouaké P.O. Box BP V18, Côte d’Ivoire
| | - Adèle Kacou N’Douba
- Training and Research Unit of Medical Sciences, Félix Houphouët-Boigny University, Abidjan P.O. Box BP 44, Côte d’Ivoire;
- Centre Hospitalier Universitaire Angré, Abidjan P.O. Box BP 1530, Côte d’Ivoire
| | - Joseph Allico Djaman
- Training and Research Unit of Biosciences, Félix Houphouët Boigny University, Abidjan P.O. Box BP V 34, Côte d’Ivoire;
| | - Mathias Herrmann
- Institute of Medical Microbiology, University Hospital Münster, 48149 Münster, Germany; (M.H.); (F.S.)
| | - Frieder Schaumburg
- Institute of Medical Microbiology, University Hospital Münster, 48149 Münster, Germany; (M.H.); (F.S.)
| | - Silke Niemann
- Institute of Medical Microbiology, University Hospital Münster, 48149 Münster, Germany; (M.H.); (F.S.)
- Correspondence: ; Tel.: +49-251-835-5369
| |
Collapse
|
46
|
Profile analysis of circRNAs in human THP-1 derived macrophages infected with intracellular Staphylococcus aureus. Microb Pathog 2022; 165:105466. [PMID: 35247499 DOI: 10.1016/j.micpath.2022.105466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 01/13/2022] [Accepted: 02/28/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Intracellular Staphylococcus aureus (S. aureus) infection is generally persistent, recurrent and difficult to treat due to the poor availability of antibiotics within macrophages cells and the lack of ideal diagnostic markers. Circular RNAs (circRNAs), with covalently closed circular structures, exists in the serum stably and is not easily degraded by nucleases. Besides, circRNAs play a pivotal in the eukaryotic regulation of genes expression and served as biomarkers in variety disease including microbial infections. However, the function of host circRNAs in intracellular S. aureus infection remains largely unclear. METHODS In this study, the circRNAs expression profile was investigated by RNA sequencing technology in both S. aureus-infected THP-1 derived macrophages and mock control cells. The differentially expressed circRNAs (DE circRNAs) with a fold-change >1.5 (p < 0.05) are analyzed using functional pathway clustering prediction. Then, RT-qPCR was performed to verify the top 2 up-regulated circRNAs in the THP-1 cell and human serum samples so as to evaluate the value of circRNAs for S. aureus diagnosis. RESULTS An intracellular survival THP-1 derived macrophages model of S. aureus infection was established. A total of 5,299 circRNAs were identified in human THP-1 derived macrophages infected with intracellular S. aureus. There were 61 DE circRNAs with a fold-change >1.5 (p < 0.05) after S. aureus infection. Among them, 22 circRNAs were up-regulated while 39 circRNAs down-regulated. GO and KEGG pathway analysis demonstrated that DE circRNAs were enriched in the processes such as Neurotrophin, Pyruvate metabolism and Notch signaling pathway. Moreover, hsa_circ_0000311 and chr13:43500472-43544806-(novel) were verified to be significantly upregulated in THP-1 derived macrophages and human serum samples between two groups. Finally, the networks of circRNA-miRNA-mRNA based on these two circRNAs were constructed respectively. CONCLUSION Our study provides the first profile analysis of host circRNAs involved in intracellular S. aureus infection, which may serve as biomarkers for S. aureus diagnosis and contribute to the understanding of S. aureus evasion mechanisms.
Collapse
|
47
|
Kember M, Grandy S, Raudonis R, Cheng Z. Non-Canonical Host Intracellular Niche Links to New Antimicrobial Resistance Mechanism. Pathogens 2022; 11:pathogens11020220. [PMID: 35215166 PMCID: PMC8876822 DOI: 10.3390/pathogens11020220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/03/2022] [Accepted: 02/05/2022] [Indexed: 12/04/2022] Open
Abstract
Globally, infectious diseases are one of the leading causes of death among people of all ages. The development of antimicrobials to treat infectious diseases has been one of the most significant advances in medical history. Alarmingly, antimicrobial resistance is a widespread phenomenon that will, without intervention, make currently treatable infections once again deadly. In an era of widespread antimicrobial resistance, there is a constant and pressing need to develop new antibacterial drugs. Unraveling the underlying resistance mechanisms is critical to fight this crisis. In this review, we summarize some emerging evidence of the non-canonical intracellular life cycle of two priority antimicrobial-resistant bacterial pathogens: Pseudomonas aeruginosa and Staphylococcus aureus. The bacterial factors that modulate this unique intracellular niche and its implications in contributing to resistance are discussed. We then briefly discuss some recent research that focused on the promises of boosting host immunity as a combination therapy with antimicrobials to eradicate these two particular pathogens. Finally, we summarize the importance of various strategies, including surveillance and vaccines, in mitigating the impacts of antimicrobial resistance in general.
Collapse
|
48
|
Investigating Morphological Changes of T-lymphocytes after Exposure with Bacterial Determinants for Early Detection of Septic Conditions. Microorganisms 2022; 10:microorganisms10020391. [PMID: 35208846 PMCID: PMC8879819 DOI: 10.3390/microorganisms10020391] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/03/2022] [Accepted: 02/05/2022] [Indexed: 12/11/2022] Open
Abstract
Sepsis is a leading cause of morbidity and mortality, annually affecting millions of people worldwide. Immediate treatment initiation is crucial to improve the outcome but despite great progress, early identification of septic patients remains a challenge. Recently, white blood cell morphology was proposed as a new biomarker for sepsis diagnosis. In this proof-of-concept study, we aimed to investigate the effect of different bacteria and their determinants on T-lymphocytes by digital holographic microscopy (DHM). We hypothesize that species- and strain-specific morphological changes occur, which may offer a new approach for early sepsis diagnosis and identification of the causative agent. Jurkat cells as a model system were exposed to different S. aureus or E. coli strains either using sterile determinants or living bacteria. Time-lapse DHM was applied to analyze cellular morphological changes. There were not only living bacteria but also membrane vesicles and sterile culture supernatant-induced changes of cell area, circularity, and mean phase contrast. Interestingly, different cellular responses occurred depending on both the species and strain of the causative bacteria. Our findings suggest that investigation of T-lymphocyte morphology might provide a promising tool for the early identification of bacterial infections and possibly discrimination between different causative agents. Distinguishing gram-positive from gram-negative infection would already offer a great benefit for the proper administration of antibiotics.
Collapse
|
49
|
Role of Implantable Drug Delivery Devices with Dual Platform Capabilities in the Prevention and Treatment of Bacterial Osteomyelitis. Bioengineering (Basel) 2022; 9:bioengineering9020065. [PMID: 35200418 PMCID: PMC8869141 DOI: 10.3390/bioengineering9020065] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 11/26/2022] Open
Abstract
As medicine advances and physicians are able to provide patients with innovative solutions, including placement of temporary or permanent medical devices that drastically improve quality of life of the patient, there is the persistent, recurring problem of chronic bacterial infection, including osteomyelitis. Osteomyelitis can manifest as a result of traumatic or contaminated wounds or implant-associated infections. This bacterial infection can persist as a result of inadequate treatment regimens or the presence of biofilm on implanted medical devices. One strategy to mitigate these concerns is the use of implantable medical devices that simultaneously act as local drug delivery devices (DDDs). This classification of device has the potential to prevent or aid in clearing chronic bacterial infection by delivering effective doses of antibiotics to the area of interest and can be engineered to simultaneously aid in tissue regeneration. This review will provide a background on bacterial infection and current therapies as well as current and prospective implantable DDDs, with a particular emphasis on local DDDs to combat bacterial osteomyelitis.
Collapse
|
50
|
Guo H, Tong Y, Cheng J, Abbas Z, Li Z, Wang J, Zhou Y, Si D, Zhang R. Biofilm and Small Colony Variants-An Update on Staphylococcus aureus Strategies toward Drug Resistance. Int J Mol Sci 2022; 23:ijms23031241. [PMID: 35163165 PMCID: PMC8835882 DOI: 10.3390/ijms23031241] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/16/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
Recently, the drawbacks arising from the overuse of antibiotics have drawn growing public attention. Among them, drug-resistance (DR) and even multidrug-resistance (MDR) pose significant challenges in clinical practice. As a representative of a DR or MDR pathogen, Staphylococcus aureus can cause diversity of infections related to different organs, and can survive or adapt to the diverse hostile environments by switching into other phenotypes, including biofilm and small colony variants (SCVs), with altered physiologic or metabolic characteristics. In this review, we briefly describe the development of the DR/MDR as well as the classical mechanisms (accumulation of the resistant genes). Moreover, we use multidimensional scaling analysis to evaluate the MDR relevant hotspots in the recent published reports. Furthermore, we mainly focus on the possible non-classical resistance mechanisms triggered by the two important alternative phenotypes of the S. aureus, biofilm and SCVs, which are fundamentally caused by the different global regulation of the S. aureus population, such as the main quorum-sensing (QS) and agr system and its coordinated regulated factors, such as the SarA family proteins and the alternative sigma factor σB (SigB). Both the biofilm and the SCVs are able to escape from the host immune response, and resist the therapeutic effects of antibiotics through the physical or the biological barriers, and become less sensitive to some antibiotics by the dormant state with the limited metabolisms.
Collapse
|