1
|
Arumugam P, Kielian T. Metabolism Shapes Immune Responses to Staphylococcus aureus. J Innate Immun 2023; 16:12-30. [PMID: 38016430 PMCID: PMC10766399 DOI: 10.1159/000535482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/21/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Staphylococcus aureus (S. aureus) is a common cause of hospital- and community-acquired infections that can result in various clinical manifestations ranging from mild to severe disease. The bacterium utilizes different combinations of virulence factors and biofilm formation to establish a successful infection, and the emergence of methicillin- and vancomycin-resistant strains introduces additional challenges for infection management and treatment. SUMMARY Metabolic programming of immune cells regulates the balance of energy requirements for activation and dictates pro- versus anti-inflammatory function. Recent investigations into metabolic adaptations of leukocytes and S. aureus during infection indicate that metabolic crosstalk plays a crucial role in pathogenesis. Furthermore, S. aureus can modify its metabolic profile to fit an array of niches for commensal or invasive growth. KEY MESSAGES Here we focus on the current understanding of immunometabolism during S. aureus infection and explore how metabolic crosstalk between the host and S. aureus influences disease outcome. We also discuss how key metabolic pathways influence leukocyte responses to other bacterial pathogens when information for S. aureus is not available. A better understanding of how S. aureus and leukocytes adapt their metabolic profiles in distinct tissue niches may reveal novel therapeutic targets to prevent or control invasive infections.
Collapse
Affiliation(s)
- Prabhakar Arumugam
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
2
|
Arciola CR, Ravaioli S, Mirzaei R, Dolzani P, Montanaro L, Daglia M, Campoccia D. Biofilms in Periprosthetic Orthopedic Infections Seen through the Eyes of Neutrophils: How Can We Help Neutrophils? Int J Mol Sci 2023; 24:16669. [PMID: 38068991 PMCID: PMC10706149 DOI: 10.3390/ijms242316669] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Despite advancements in our knowledge of neutrophil responses to planktonic bacteria during acute inflammation, much remains to be elucidated on how neutrophils deal with bacterial biofilms in implant infections. Further complexity transpires from the emerging findings on the role that biomaterials play in conditioning bacterial adhesion, the variety of biofilm matrices, and the insidious measures that biofilm bacteria devise against neutrophils. Thus, grasping the entirety of neutrophil-biofilm interactions occurring in periprosthetic tissues is a difficult goal. The bactericidal weapons of neutrophils consist of the following: ready-to-use antibacterial proteins and enzymes stored in granules; NADPH oxidase-derived reactive oxygen species (ROS); and net-like structures of DNA, histones, and granule proteins, which neutrophils extrude to extracellularly trap pathogens (the so-called NETs: an allusive acronym for "neutrophil extracellular traps"). Neutrophils are bactericidal (and therefore defensive) cells endowed with a rich offensive armamentarium through which, if frustrated in their attempts to engulf and phagocytose biofilms, they can trigger the destruction of periprosthetic bone. This study speculates on how neutrophils interact with biofilms in the dramatic scenario of implant infections, also considering the implications of this interaction in view of the design of new therapeutic strategies and functionalized biomaterials, to help neutrophils in their arduous task of managing biofilms.
Collapse
Affiliation(s)
- Carla Renata Arciola
- Laboratory of Immunorheumatology and Tissue Regeneration, Laboratory of Pathology of Implant Infections, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy
| | - Stefano Ravaioli
- Laboratorio di Patologia delle Infezioni Associate all’Impianto, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (S.R.); (D.C.)
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Laboratory, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Iran;
| | - Paolo Dolzani
- Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
| | - Lucio Montanaro
- Laboratory of Immunorheumatology and Tissue Regeneration, Laboratory of Pathology of Implant Infections, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy;
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via San Giacomo 14, 40126 Bologna, Italy
| | - Maria Daglia
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Naples, Italy;
| | - Davide Campoccia
- Laboratorio di Patologia delle Infezioni Associate all’Impianto, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (S.R.); (D.C.)
| |
Collapse
|
3
|
Fantone KM, Goldberg JB, Stecenko AA, Rada B. Sputum from People with Cystic Fibrosis Reduces the Killing of Methicillin-Resistant Staphylococcus aureus by Neutrophils and Diminishes Phagosomal Production of Reactive Oxygen Species. Pathogens 2023; 12:1148. [PMID: 37764956 PMCID: PMC10538153 DOI: 10.3390/pathogens12091148] [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: 08/04/2023] [Revised: 08/23/2023] [Accepted: 08/27/2023] [Indexed: 09/29/2023] Open
Abstract
Cystic fibrosis (CF) airway disease is characterized by chronic polymicrobial infections and an infiltration of neutrophils (PMNs). Staphylococcus aureus has been the most prevalent respiratory pathogen in CF. In particular, methicillin-resistant S. aureus (MRSA) represents a huge clinical burden in CF due to its association with lung disease and increased resistance to antibiotics. In CF, PMNs are unable to kill and clear MRSA. The reason for this remains largely unknown. Our study found that CF PMNs are as equally capable of killing MRSA as healthy PMNs. We show that the CF sputum, however, significantly impairs the ability of human PMNs to kill CF MRSA isolates. In the absence of CF sputum, PMNs kill MRSA via intracellular mechanisms mediated by phagocytosis, rather than extracellular mechanisms via NET formation. CF sputum does not affect the phagocytosis of MRSA via healthy or CF PMNs. Our results demonstrate that CF sputum exposure impairs phagosomal levels of reactive oxygen species (ROS) in MRSA-phagocytosing PMNs. While phagosomal co-localizations of MRSA with primary granule markers, myeloperoxidase and cathepsin D, were significantly reduced upon CF sputum exposure, that of a third azurophilic granule marker, neutrophil elastase, remained unaffected. This suggests that CF sputum does not compromise the fusion of primary granules with phagosomes but diminishes phagosomal ROS levels via another, likely more specific, mechanism. Overall, we identified the airway environment as an important factor that restricts neutrophils' oxidative microbicidal activities in CF against MRSA. These results deliver new details of the complex host-pathogen interactions present in the CF lung.
Collapse
Affiliation(s)
- Kayla M. Fantone
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA;
| | - Joanna B. Goldberg
- Division of Pulmonology, Asthma, Cystic Fibrosis and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30602, USA; (J.B.G.); (A.A.S.)
| | - Arlene A. Stecenko
- Division of Pulmonology, Asthma, Cystic Fibrosis and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30602, USA; (J.B.G.); (A.A.S.)
| | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
4
|
Chen Y, Liu Z, Lin Z, Lu M, Fu Y, Liu G, Yu B. The effect of Staphylococcus aureus on innate and adaptive immunity and potential immunotherapy for S. aureus-induced osteomyelitis. Front Immunol 2023; 14:1219895. [PMID: 37744377 PMCID: PMC10517662 DOI: 10.3389/fimmu.2023.1219895] [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: 05/09/2023] [Accepted: 08/18/2023] [Indexed: 09/26/2023] Open
Abstract
Osteomyelitis is a chronic inflammatory bone disease caused by infection of open fractures or post-operative implants. Particularly in patients with open fractures, the risk of osteomyelitis is greatly increased as the soft tissue damage and bacterial infection are often more severe. Staphylococcus aureus, one of the most common pathogens of osteomyelitis, disrupts the immune response through multiple mechanisms, such as biofilm formation, virulence factor secretion, and metabolic pattern alteration, which attenuates the effectiveness of antibiotics and surgical debridement toward osteomyelitis. In osteomyelitis, immune cells such as neutrophils, macrophages and T cells are activated in response to pathogenic bacteria invasion with excessive inflammatory factor secretion, immune checkpoint overexpression, and downregulation of immune pathway transcription factors, which enhances osteoclastogenesis and results in bone destruction. Therefore, the study of the mechanisms of abnormal immunity will be a new breakthrough in the treatment of osteomyelitis.
Collapse
Affiliation(s)
- Yingqi Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Zixian Liu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Zexin Lin
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Mincheng Lu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Yong Fu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
- Trauma Center, Department of Orthopaedic Trauma, The Second Affiliated Hospital of Hengyang Medical College, South China University, Hengyang, China
| | - Guanqiao Liu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| |
Collapse
|
5
|
Tomlinson KL, Riquelme SA, Baskota SU, Drikic M, Monk IR, Stinear TP, Lewis IA, Prince AS. Staphylococcus aureus stimulates neutrophil itaconate production that suppresses the oxidative burst. Cell Rep 2023; 42:112064. [PMID: 36724077 PMCID: PMC10387506 DOI: 10.1016/j.celrep.2023.112064] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 12/01/2022] [Accepted: 01/18/2023] [Indexed: 02/02/2023] Open
Abstract
Neutrophils are critical in the host defense against Staphylococcus aureus, a major human pathogen. However, even in the setting of a robust neutrophil response, S. aureus can evade immune clearance. Here, we demonstrate that S. aureus impairs neutrophil function by triggering the production of the anti-inflammatory metabolite itaconate. The enzyme that synthesizes itaconate, Irg1, is selectively expressed in neutrophils during S. aureus pneumonia. Itaconate inhibits neutrophil glycolysis and oxidative burst, which impairs survival and bacterial killing. In a murine pneumonia model, neutrophil Irg1 expression protects the lung from excessive inflammation but compromises bacterial clearance. S. aureus is thus able to evade the innate immune response by targeting neutrophil metabolism and inducing the production of the anti-inflammatory metabolite itaconate.
Collapse
Affiliation(s)
- Kira L Tomlinson
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | | | | | - Marija Drikic
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Ian R Monk
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Timothy P Stinear
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Ian A Lewis
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Alice S Prince
- Department of Pediatrics, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
6
|
NOD2 Agonism Counter-Regulates Human Type 2 T Cell Functions in Peripheral Blood Mononuclear Cell Cultures: Implications for Atopic Dermatitis. Biomolecules 2023; 13:biom13020369. [PMID: 36830738 PMCID: PMC9953199 DOI: 10.3390/biom13020369] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/31/2023] [Accepted: 02/03/2023] [Indexed: 02/18/2023] Open
Abstract
Atopic dermatitis (AD) is known as a skin disease; however, T cell immunopathology found in blood is associated with its severity. Skin Staphylococcus aureus (S. aureus) and associated host-pathogen dynamics are important to chronic T helper 2 (Th2)-dominated inflammation in AD, yet they remain poorly understood. This study sought to investigate the effects of S. aureus-derived molecules and skin alarmins on human peripheral blood mononuclear cells, specifically testing Th2-type cells, cytokines, and chemokines known to be associated with AD. We first show that six significantly elevated Th2-related chemokine biomarkers distinguish blood from adult AD patients compared to healthy controls ex vivo; in addition, TARC/CCL17, LDH, and PDGF-AA/AB correlated significantly with disease severity. We then demonstrate that these robust AD-associated biomarkers, as well as associated type 2 T cell functions, are readily reproduced from healthy blood mononuclear cells exposed to the alarmin TSLP and the S. aureus superantigen SEB in a human in vitro model, including IL-13, IL-5, and TARC secretion as well as OX-40-expressing activated memory T cells. We further show that the agonism of nucleotide-binding oligomerization domain-containing protein (NOD)2 inhibits this IL-13 secretion and memory Th2 and Tc2 cell functional activation while inducing significantly increased pSTAT3 and IL-6, both critical for Th17 cell responses. These findings identify NOD2 as a potential regulator of type 2 immune responses in humans and highlight its role as an endogenous inhibitor of pathogenic IL-13 that may open avenues for its therapeutic targeting in AD.
Collapse
|
7
|
Scutera S, Sparti R, Comini S, Menotti F, Musso T, Cuffini AM, Allizond V, Banche G. Dalbavancin Boosts the Ability of Neutrophils to Fight Methicillin-Resistant Staphylococcus aureus. Int J Mol Sci 2023; 24:ijms24032541. [PMID: 36768864 PMCID: PMC9917267 DOI: 10.3390/ijms24032541] [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: 12/30/2022] [Revised: 01/18/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Polymorphonuclear leukocytes (PMNs) are the most important cell type involved in the early nonspecific host response to bacterial pathogens. Staphylococcus aureus has evolved mechanisms to evade immune responses that contribute to its persistence in PMNs, and acquired resistance to several antimicrobials. Additionally, methicillin-resistant S. aureus (MRSA) is one of the most common causes of acute bacterial skin and skin-structure infections (ABSSSIs). Dalbavancin (DBV), a lipoglycopeptide, is indicated for the treatment of ABSSSIs, and has a broad spectrum of action against most microorganisms. Here, we sought to determine the effect of DBV on the neutrophil killing of MRSA and its potential immunomodulating activity. Our results revealed that DBV boosts MRSA killing by acting on both bacteria and PMNs. DBV pre-treatment of PMNs did not change the respiratory burst or degranulation, while an increased trend in neutrophil extracellular traps-associated elastase and in the production of TNFα and CXCL8 was revealed. In parallel, DBV caused a delay in the apoptosis of MRSA-infected neutrophils. In conclusion, we demonstrated a cooperative effect between the antimicrobial properties of PMNs and DBV, thus owing to their immunomodulatory activity. In the choice of the treatment management of serious S. aureus infections, DBV should be considered as an outstanding option since it reinforces PMNs pathogen clearance capability by exerting its effect directly, not only on MRSA but also on neutrophils.
Collapse
|
8
|
Ashby LV, Springer R, Loi VV, Antelmann H, Hampton MB, Kettle AJ, Dickerhof N. Oxidation of bacillithiol during killing of Staphylococcus aureus USA300 inside neutrophil phagosomes. J Leukoc Biol 2022; 112:591-605. [PMID: 35621076 PMCID: PMC9796752 DOI: 10.1002/jlb.4hi1021-538rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 03/29/2022] [Indexed: 01/07/2023] Open
Abstract
Targeting immune evasion tactics of pathogenic bacteria may hold the key to treating recalcitrant bacterial infections. Staphylococcus aureus produces bacillithiol (BSH), its major low-molecular-weight thiol, which is thought to protect this opportunistic human pathogen against the bombardment of oxidants inside neutrophil phagosomes. Here, we show that BSH was oxidized when human neutrophils phagocytosed S. aureus, but provided limited protection to the bacteria. We used mass spectrometry to measure the oxidation of BSH upon exposure of S. aureus USA300 to either a bolus of hypochlorous acid (HOCl) or a flux generated by the neutrophil enzyme myeloperoxidase. Oxidation of BSH and loss of bacterial viability were strongly correlated (r = 0.99, p < 0.001). BSH was fully oxidized after exposure of S. aureus to lethal doses of HOCl. However, there was no relationship between the initial BSH levels and the dose of HOCl required for bacterial killing. In contrast to the HOCl systems, only 50% of total BSH was oxidized when neutrophils killed the majority of phagocytosed bacteria. Oxidation of BSH was decreased upon inhibition of myeloperoxidase, implicating HOCl in phagosomal BSH oxidation. A BSH-deficient S. aureus USA300 mutant was slightly more susceptible to treatment with either HOCl or ammonia chloramine, or to killing within neutrophil phagosomes. Collectively, our data show that myeloperoxidase-derived oxidants react with S. aureus inside neutrophil phagosomes, leading to partial BSH oxidation, and contribute to bacterial killing. However, BSH offers only limited protection against the neutrophil's multifaceted killing mechanisms.
Collapse
Affiliation(s)
- Louisa V Ashby
- Centre for Free Radical Research, Department of Pathology and Biomedical ScienceUniversity of Otago ChristchurchChristchurchNew Zealand
| | - Reuben Springer
- Centre for Free Radical Research, Department of Pathology and Biomedical ScienceUniversity of Otago ChristchurchChristchurchNew Zealand
| | - Vu Van Loi
- Freie Universität Berlin, Department of Biology, Chemistry, PharmacyInstitute of Biology‐MicrobiologyBerlinGermany
| | - Haike Antelmann
- Freie Universität Berlin, Department of Biology, Chemistry, PharmacyInstitute of Biology‐MicrobiologyBerlinGermany
| | - Mark B Hampton
- Centre for Free Radical Research, Department of Pathology and Biomedical ScienceUniversity of Otago ChristchurchChristchurchNew Zealand
| | - Anthony J Kettle
- Centre for Free Radical Research, Department of Pathology and Biomedical ScienceUniversity of Otago ChristchurchChristchurchNew Zealand
| | - Nina Dickerhof
- Centre for Free Radical Research, Department of Pathology and Biomedical ScienceUniversity of Otago ChristchurchChristchurchNew Zealand
| |
Collapse
|
9
|
Chee Wezen X, Chandran A, Eapen RS, Waters E, Bricio-Moreno L, Tosi T, Dolan S, Millership C, Kadioglu A, Gründling A, Itzhaki LS, Welch M, Rahman T. Structure-Based Discovery of Lipoteichoic Acid Synthase Inhibitors. J Chem Inf Model 2022; 62:2586-2599. [PMID: 35533315 PMCID: PMC9131456 DOI: 10.1021/acs.jcim.2c00300] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Indexed: 01/20/2023]
Abstract
Lipoteichoic acid synthase (LtaS) is a key enzyme for the cell wall biosynthesis of Gram-positive bacteria. Gram-positive bacteria that lack lipoteichoic acid (LTA) exhibit impaired cell division and growth defects. Thus, LtaS appears to be an attractive antimicrobial target. The pharmacology around LtaS remains largely unexplored with only two small-molecule LtaS inhibitors reported, namely "compound 1771" and the Congo red dye. Structure-based drug discovery efforts against LtaS remain unattempted due to the lack of an inhibitor-bound structure of LtaS. To address this, we combined the use of a molecular docking technique with molecular dynamics (MD) simulations to model a plausible binding mode of compound 1771 to the extracellular catalytic domain of LtaS (eLtaS). The model was validated using alanine mutagenesis studies combined with isothermal titration calorimetry. Additionally, lead optimization driven by our computational model resulted in an improved version of compound 1771, namely, compound 4 which showed greater affinity for binding to eLtaS than compound 1771 in biophysical assays. Compound 4 reduced LTA production in S. aureus dose-dependently, induced aberrant morphology as seen for LTA-deficient bacteria, and significantly reduced bacteria titers in the lung of mice infected with S. aureus. Analysis of our MD simulation trajectories revealed the possible formation of a transient cryptic pocket in eLtaS. Virtual screening (VS) against the cryptic pocket led to the identification of a new class of inhibitors that could potentiate β-lactams against methicillin-resistant S. aureus. Our overall workflow and data should encourage further drug design campaign against LtaS. Finally, our work reinforces the importance of considering protein conformational flexibility to a successful VS endeavor.
Collapse
Affiliation(s)
- Xavier Chee Wezen
- Science
Program, School of Chemical Engineering and Science, Faculty of Engineering,
Computing and Science, Swinburne University
of Technology Sarawak, Kuching 93350, Malaysia
| | - Aneesh Chandran
- Department
of Biotechnology & Microbiology, Kannur
University, Kannur 670 661, Kerala, India
| | | | - Elaine Waters
- Department
of Clinical Infection Microbiology and Immunology, Institute of Infection
and Global Health, University of Liverpool, Liverpool L69 7BE, U.K.
| | - Laura Bricio-Moreno
- Department
of Clinical Infection Microbiology and Immunology, Institute of Infection
and Global Health, University of Liverpool, Liverpool L69 7BE, U.K.
| | - Tommaso Tosi
- Section
of Molecular Microbiology and MRC Centre for Molecular Bacteriology
and Infection, Imperial College London, London SW7 2AZ, U.K.
| | - Stephen Dolan
- Department
of Biochemistry, University of Cambridge, Cambridge CB2 1QW, U.K.
| | - Charlotte Millership
- Section
of Molecular Microbiology and MRC Centre for Molecular Bacteriology
and Infection, Imperial College London, London SW7 2AZ, U.K.
| | - Aras Kadioglu
- Department
of Clinical Infection Microbiology and Immunology, Institute of Infection
and Global Health, University of Liverpool, Liverpool L69 7BE, U.K.
| | - Angelika Gründling
- Section
of Molecular Microbiology and MRC Centre for Molecular Bacteriology
and Infection, Imperial College London, London SW7 2AZ, U.K.
| | - Laura S. Itzhaki
- Department
of PharmacologyUniversity of CambridgeCambridgeCB2 1PDU.K.
| | - Martin Welch
- Department
of Biochemistry, University of Cambridge, Cambridge CB2 1QW, U.K.
| | - Taufiq Rahman
- Department
of PharmacologyUniversity of CambridgeCambridgeCB2 1PDU.K.
| |
Collapse
|
10
|
Tang Q, Precit MR, Thomason MK, Blanc SF, Ahmed-Qadri F, McFarland AP, Wolter DJ, Hoffman LR, Woodward JJ. Thymidine starvation promotes c-di-AMP-dependent inflammation during pathogenic bacterial infection. Cell Host Microbe 2022; 30:961-974.e6. [PMID: 35439435 DOI: 10.1016/j.chom.2022.03.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 02/01/2022] [Accepted: 03/23/2022] [Indexed: 11/03/2022]
Abstract
Antimicrobials can impact bacterial physiology and host immunity with negative treatment outcomes. Extensive exposure to antifolate antibiotics promotes thymidine-dependent Staphylococcus aureus small colony variants (TD-SCVs), commonly associated with worse clinical outcomes. We show that antibiotic-mediated disruption of thymidine synthesis promotes elevated levels of the bacterial second messenger cyclic di-AMP (c-di-AMP), consequently inducing host STING activation and inflammation. An initial antibiotic screen in Firmicutes revealed that c-di-AMP production was largely driven by antifolate antibiotics targeting dihydrofolate reductase (DHFR), which promotes folate regeneration required for thymidine biosynthesis. Additionally, TD-SCVs exhibited excessive c-di-AMP production and STING activation in a thymidine-dependent manner. Murine lung infection with TD-SCVs revealed STING-dependent elevation of proinflammatory cytokines, causing higher airway neutrophil infiltration and activation compared with normal-colony S. aureus and hemin-dependent SCVs. Collectively, our results suggest that thymidine metabolism disruption in Firmicutes leads to elevated c-di-AMP-mediated STING-dependent inflammation, with potential impacts on antibiotic usage and infection outcomes.
Collapse
Affiliation(s)
- Qing Tang
- Department of Microbiology, University of Washington, Seattle, WA 98105, USA
| | - Mimi R Precit
- Department of Microbiology, University of Washington, Seattle, WA 98105, USA
| | - Maureen K Thomason
- Department of Microbiology, University of Washington, Seattle, WA 98105, USA
| | - Sophie F Blanc
- Department of Microbiology, University of Washington, Seattle, WA 98105, USA
| | - Fariha Ahmed-Qadri
- Department of Microbiology, University of Washington, Seattle, WA 98105, USA
| | - Adelle P McFarland
- Department of Microbiology, University of Washington, Seattle, WA 98105, USA
| | - Daniel J Wolter
- Department of Pediatrics, University of Washington, Seattle, WA 98105, USA; Pulmonary and Sleep Medicine, Seattle Children's Hospital, Seattle, WA 98105, USA
| | - Lucas R Hoffman
- Department of Microbiology, University of Washington, Seattle, WA 98105, USA; Department of Pediatrics, University of Washington, Seattle, WA 98105, USA; Pulmonary and Sleep Medicine, Seattle Children's Hospital, Seattle, WA 98105, USA
| | - Joshua J Woodward
- Department of Microbiology, University of Washington, Seattle, WA 98105, USA.
| |
Collapse
|
11
|
Pasman R, Krom BP, Zaat SAJ, Brul S. The Role of the Oral Immune System in Oropharyngeal Candidiasis-Facilitated Invasion and Dissemination of Staphylococcus aureus. FRONTIERS IN ORAL HEALTH 2022; 3:851786. [PMID: 35464779 PMCID: PMC9021398 DOI: 10.3389/froh.2022.851786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/25/2022] [Indexed: 11/13/2022] Open
Abstract
Candida albicans and Staphylococcus aureus account for most invasive fungal and bacterial bloodstream infections (BSIs), respectively. However, the initial point of invasion responsible for S. aureus BSIs is often unclear. Recently, C. albicans has been proposed to mediate S. aureus invasion of immunocompromised hosts during co-colonization of oral mucosal surfaces. The status of the oral immune system crucially contributes to this process in two distinct ways: firstly, by allowing invasive C. albicans growth during dysfunction of extra-epithelial immunity, and secondly following invasion by some remaining function of intra-epithelial immunity. Immunocompromised individuals at risk of developing invasive oral C. albicans infections could, therefore, also be at risk of contracting concordant S. aureus BSIs. Considering the crucial contribution of both oral immune function and dysfunction, the aim of this review is to provide an overview of relevant aspects of intra and extra-epithelial oral immunity and discuss predominant immune deficiencies expected to facilitate C. albicans induced S. aureus BSIs.
Collapse
Affiliation(s)
- Raymond Pasman
- Department of Molecular Biology and Microbial Food Safety, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Bastiaan P. Krom
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Sebastian A. J. Zaat
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Stanley Brul
- Department of Molecular Biology and Microbial Food Safety, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
- *Correspondence: Stanley Brul
| |
Collapse
|
12
|
A Factor H-Fc fusion protein increases complement-mediated opsonophagocytosis and killing of community associated methicillin-resistant Staphylococcus aureus. PLoS One 2022; 17:e0265774. [PMID: 35324969 PMCID: PMC8946749 DOI: 10.1371/journal.pone.0265774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 03/07/2022] [Indexed: 11/19/2022] Open
Abstract
Staphylococcus aureus employs a multitude of immune-evasive tactics to circumvent host defenses including the complement system, a component of innate immunity central to controlling bacterial infections. With antibiotic resistance becoming increasingly common, there is a dire need for novel therapies. Previously, we have shown that S. aureus binds the complement regulator factor H (FH) via surface protein SdrE to inhibit complement. To address the need for novel therapeutics and take advantage of the FH:SdrE interaction, we examined the effect of a fusion protein comprised of the SdrE-interacting domain of FH coupled with IgG Fc on complement-mediated opsonophagocytosis and bacterial killing of community associated methicillin-resistant S. aureus. S. aureus bound significantly more FH-Fc compared to Fc-control proteins and FH-Fc competed with serum FH for S. aureus binding. FH-Fc treatment increased C3-fragment opsonization of S. aureus for both C3b and iC3b, and boosted generation of the anaphylatoxin C5a. In 5 and 10% serum, FH-Fc treatment significantly increased S. aureus killing by polymorphonuclear cells. This anti-staphylococcal effect was evident in 75% (3/4) of clinical isolates tested. This study demonstrates that FH-Fc fusion proteins have the potential to mitigate the protective effects of bound serum FH rendering S. aureus more vulnerable to the host immune system. Thus, we report the promise of virulence-factor-targeted fusion-proteins as an avenue for prospective anti-staphylococcal therapeutic development.
Collapse
|
13
|
Boero E, Cruz AR, Pansegrau W, Giovani C, Rooijakkers SHM, van Kessel KPM, van Strijp JAG, Bagnoli F, Manetti AGO. Natural Human Immunity Against Staphylococcal Protein A Relies on Effector Functions Triggered by IgG3. Front Immunol 2022; 13:834711. [PMID: 35359919 PMCID: PMC8963248 DOI: 10.3389/fimmu.2022.834711] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/17/2022] [Indexed: 12/02/2022] Open
Abstract
Staphylococcal protein A (SpA) is a multifunctional, highly conserved virulence factor of Staphylococcus aureus. By binding the Fc portion of all human IgG subclasses apart from IgG3, SpA interferes with antibody and complement deposition on the bacterial surface, impairing staphylococcal clearance by phagocytosis. Because of its anti-opsonic properties, SpA is not investigated as a surface antigen to mediate bacterial phagocytosis. Herein we investigate human sera for the presence of SpA-opsonizing antibodies. The screening revealed that sera containing IgG3 against SpA were able to correctly opsonize the target and drive Fcγ receptor-mediated interactions and phagocytosis. We demonstrated that IgG3 Fc is significantly more efficient in inducing phagocytosis of SpA-expressing S. aureus as compared to IgG1 Fc in an assay resembling physiological conditions. Furthermore, we show that the capacity of SpA antibodies to induce phagocytosis depends on the specific epitope recognized by the IgGs on SpA molecules. Overall, our results suggest that anti-SpA IgG3 antibodies could favor the anti-staphylococcal response in humans, paving the way towards the identification of a correlate of protection against staphylococcal infections.
Collapse
Affiliation(s)
- Elena Boero
- GSK, Siena, Italy
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Ana Rita Cruz
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | | | | | - Suzan H. M. Rooijakkers
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Kok P. M. van Kessel
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Jos A. G. van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | | | | |
Collapse
|
14
|
Wang L, Letsiou E, Wang H, Belvitch P, Meliton LN, Brown ME, Bandela M, Chen J, Garcia JGN, Dudek SM. MRSA-induced endothelial permeability and acute lung injury are attenuated by FTY720 S-phosphonate. Am J Physiol Lung Cell Mol Physiol 2022; 322:L149-L161. [PMID: 35015568 PMCID: PMC8794017 DOI: 10.1152/ajplung.00100.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Disruption of the lung endothelial barrier is a hallmark of acute respiratory distress syndrome (ARDS), for which no effective pharmacologic treatments exist. Prior work has demonstrated that FTY720 S-phosphonate (Tys), an analog of sphingosine-1-phosphate (S1P) and FTY720, exhibits potent endothelial cell (EC) barrier protective properties. In this study, we investigated the in vitro and in vivo efficacy of Tys against methicillin-resistant Staphylococcus aureus (MRSA), a frequent bacterial cause of ARDS. Tys-protected human lung EC from barrier disruption induced by heat-killed MRSA (HK-MRSA) or staphylococcal α-toxin and attenuated MRSA-induced cytoskeletal changes associated with barrier disruption, including actin stress fiber formation and loss of peripheral VE-cadherin and cortactin. Tys-inhibited Rho and myosin light chain (MLC) activation after MRSA and blocked MRSA-induced NF-κB activation and release of the proinflammatory cytokines, IL-6 and IL-8. In vivo, intratracheal administration of live MRSA in mice caused significant vascular leakage and leukocyte infiltration into the alveolar space. Pre- or posttreatment with Tys attenuated MRSA-induced lung permeability and levels of alveolar neutrophils. Posttreatment with Tys significantly reduced levels of bronchoalveolar lavage (BAL) VCAM-1 and plasma IL-6 and KC induced by MRSA. Dynamic intravital imaging of mouse lungs demonstrated Tys attenuation of HK-MRSA-induced interstitial edema and neutrophil infiltration into lung tissue. Tys did not directly inhibit MRSA growth or viability in vitro. In conclusion, Tys inhibits lung EC barrier disruption and proinflammatory signaling induced by MRSA in vitro and attenuates acute lung injury induced by MRSA in vivo. These results support the potential utility of Tys as a novel ARDS therapeutic strategy.
Collapse
Affiliation(s)
- Lichun Wang
- 1Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Eleftheria Letsiou
- 1Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Huashan Wang
- 1Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Patrick Belvitch
- 1Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Lucille N. Meliton
- 1Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Mary E. Brown
- 2Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota
| | - Mounica Bandela
- 1Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Jiwang Chen
- 1Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | | | - Steven M. Dudek
- 1Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
15
|
Contribution of Coagulase and Its Regulator SaeRS to Lethality of CA-MRSA 923 Bacteremia. Pathogens 2021; 10:pathogens10111396. [PMID: 34832552 PMCID: PMC8623987 DOI: 10.3390/pathogens10111396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/18/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
Coagulase is a critical factor for distinguishing Staphylococcus aureus and coagulase-negative Staphylococcus. Our previous studies demonstrated that the null mutation of coagulase (coa) or its direct regulator, SaeRS, significantly enhanced the ability of S. aureus (CA-MRSA 923) to survive in human blood in vitro. This led us to further investigate the role of coagulase and its direct regulator, SaeRS, in the pathogenicity of CA-MRSA 923 in bacteremia during infection. In this study, we found that the null mutation of coa significantly decreased the mortality of CA-MRSA 923; moreover, the single null mutation of saeRS and the double deletion of coa/saeRS abolished the virulence of CA-MRSA 923. Moreover, the mice infected with either the saeRS knockout or the coa/saeRS double knockout mutant exhibited fewer histological lesions and less neutrophils infiltration in the infected kidneys compared to those infected with the coa knockout mutant or their parental control. Furthermore, we examined the impact of coa and saeRS on bacterial survival in vitro. The null mutation of coa had no impact on bacterial survival in mice blood, whereas the deletion mutation of saeRS or coa/saeRS significantly enhanced bacterial survival in mice blood. These data indicate that SaeRS plays a key role in the lethality of CA-MRSA 923 bacteremia, and that coagulase is one of the important virulence factors that is regulated by SaeRS and contributes to the pathogenicity of CA-MRSA 923.
Collapse
|
16
|
Haikal C, Ortigosa-Pascual L, Najarzadeh Z, Bernfur K, Svanbergsson A, Otzen DE, Linse S, Li JY. The Bacterial Amyloids Phenol Soluble Modulins from Staphylococcus aureus Catalyze Alpha-Synuclein Aggregation. Int J Mol Sci 2021; 22:11594. [PMID: 34769023 PMCID: PMC8584152 DOI: 10.3390/ijms222111594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 12/02/2022] Open
Abstract
Aggregated α-synuclein (α-syn) is the main constituent of Lewy bodies, which are a pathological hallmark of Parkinson's disease (PD). Environmental factors are thought to be potential triggers capable of initiating the aggregation of the otherwise monomeric α-syn. Braak's seminal work redirected attention to the intestine and recent reports of dysbiosis have highlighted the potential causative role of the microbiome in the initiation of pathology of PD. Staphylococcus aureus is a bacterium carried by 30-70% of the general population. It has been shown to produce functional amyloids, called phenol soluble modulins (PSMαs). Here, we studied the kinetics of α-syn aggregation under quiescent conditions in the presence or absence of four different PSMα peptides and observed a remarkable shortening of the lag phase in their presence. Whereas pure α-syn monomer did not aggregate up to 450 h after initiation of the experiment in neither neutral nor mildly acidic buffer, the addition of different PSMα peptides resulted in an almost immediate increase in the Thioflavin T (ThT) fluorescence. Despite similar peptide sequences, the different PSMα peptides displayed distinct effects on the kinetics of α-syn aggregation. Kinetic analyses of the data suggest that all four peptides catalyze α-syn aggregation through heterogeneous primary nucleation. The immunogold electron microscopic analyses showed that the aggregates were fibrillar and composed of α-syn. In addition of the co-aggregated materials to a cell model expressing the A53T α-syn variant fused to GFP was found to catalyze α-syn aggregation and phosphorylation in the cells. Our results provide evidence of a potential trigger of synucleinopathies and could have implications for the prevention of the diseases.
Collapse
Affiliation(s)
- Caroline Haikal
- Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (C.H.); (A.S.)
| | - Lei Ortigosa-Pascual
- Department of Biochemistry and Structural Biology, Lund University, 22100 Lund, Sweden; (L.O.-P.); (K.B.); (S.L.)
| | - Zahra Najarzadeh
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark; (Z.N.); (D.E.O.)
| | - Katja Bernfur
- Department of Biochemistry and Structural Biology, Lund University, 22100 Lund, Sweden; (L.O.-P.); (K.B.); (S.L.)
| | - Alexander Svanbergsson
- Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (C.H.); (A.S.)
| | - Daniel E. Otzen
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark; (Z.N.); (D.E.O.)
| | - Sara Linse
- Department of Biochemistry and Structural Biology, Lund University, 22100 Lund, Sweden; (L.O.-P.); (K.B.); (S.L.)
| | - Jia-Yi Li
- Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (C.H.); (A.S.)
- Health Sciences Institute, China Medical University, Shenyang 110112, China
| |
Collapse
|
17
|
Sun J, Uchiyama S, Olson J, Morodomi Y, Cornax I, Ando N, Kohno Y, Kyaw MMT, Aguilar B, Haste NM, Kanaji S, Kanaji T, Rose WE, Sakoulas G, Marth JD, Nizet V. Repurposed drugs block toxin-driven platelet clearance by the hepatic Ashwell-Morell receptor to clear Staphylococcus aureus bacteremia. Sci Transl Med 2021; 13:13/586/eabd6737. [PMID: 33762439 DOI: 10.1126/scitranslmed.abd6737] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 03/01/2021] [Indexed: 12/13/2022]
Abstract
Staphylococcus aureus (SA) bloodstream infections cause high morbidity and mortality (20 to 30%) despite modern supportive care. In a human bacteremia cohort, we found that development of thrombocytopenia was correlated to increased mortality and increased α-toxin expression by the pathogen. Platelet-derived antibacterial peptides are important in bloodstream defense against SA, but α-toxin decreased platelet viability, induced platelet sialidase to cause desialylation of platelet glycoproteins, and accelerated platelet clearance by the hepatic Ashwell-Morell receptor (AMR). Ticagrelor (Brilinta), a commonly prescribed P2Y12 receptor inhibitor used after myocardial infarction, blocked α-toxin-mediated platelet injury and resulting thrombocytopenia, thereby providing protection from lethal SA infection in a murine intravenous challenge model. Genetic deletion or pharmacological inhibition of AMR stabilized platelet counts and enhanced resistance to SA infection, and the anti-influenza sialidase inhibitor oseltamivir (Tamiflu) provided similar therapeutic benefit. Thus, a "toxin-platelet-AMR" regulatory pathway plays a critical role in the pathogenesis of SA bloodstream infection, and its elucidation provides proof of concept for repurposing two commonly prescribed drugs as adjunctive therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Josh Sun
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA.,Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA.,Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA 92093, USA
| | - Satoshi Uchiyama
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - Joshua Olson
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - Yosuke Morodomi
- Department of Molecular Medicine, MERU-Roon Research Center on Vascular Biology, Scripps Research, La Jolla, CA 92037, USA
| | - Ingrid Cornax
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - Nao Ando
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - Yohei Kohno
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - May M T Kyaw
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - Bernice Aguilar
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA
| | - Nina M Haste
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA.,Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA.,Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA 92093, USA
| | - Sachiko Kanaji
- Department of Molecular Medicine, MERU-Roon Research Center on Vascular Biology, Scripps Research, La Jolla, CA 92037, USA
| | - Taisuke Kanaji
- Department of Molecular Medicine, MERU-Roon Research Center on Vascular Biology, Scripps Research, La Jolla, CA 92037, USA
| | - Warren E Rose
- School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
| | - George Sakoulas
- Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA
| | - Jamey D Marth
- Center for Nanomedicine, UC Santa Barbara, Santa Barbara, CA 93106, USA.,Sanford Burnham Prebys Medical Discovery Institute, UC Santa Barbara, Santa Barbara, CA 93106, USA
| | - Victor Nizet
- Biomedical Sciences Graduate Program, UC San Diego, La Jolla, CA 92093, USA. .,Department of Pediatrics, UC San Diego, La Jolla, CA 92093, USA.,Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
18
|
Vozza EG, Mulcahy ME, McLoughlin RM. Making the Most of the Host; Targeting the Autophagy Pathway Facilitates Staphylococcus aureus Intracellular Survival in Neutrophils. Front Immunol 2021; 12:667387. [PMID: 34220813 PMCID: PMC8242348 DOI: 10.3389/fimmu.2021.667387] [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: 02/12/2021] [Accepted: 05/28/2021] [Indexed: 12/21/2022] Open
Abstract
The success of Staphylococcus aureus as a human commensal and an opportunistic pathogen relies on its ability to adapt to several niches within the host. The innate immune response plays a key role in protecting the host against S. aureus infection; however, S. aureus adeptness at evading the innate immune system is indisputably evident. The “Trojan horse” theory has been postulated to describe a mechanism by which S. aureus takes advantage of phagocytes as a survival niche within the host to facilitate dissemination of S. aureus to secondary sites during systemic infection. Several studies have determined that S. aureus can parasitize both professional and non-professional phagocytes by manipulating the host autophagy pathway in order to create an intracellular survival niche. Neutrophils represent a critical cell type in S. aureus infection as demonstrated by the increased risk of infection among patients with congenital neutrophil disorders. However, S. aureus has been repeatedly shown to survive intracellularly within neutrophils with evidence now supporting a pathogenic role of host autophagy. By manipulating this pathway, S. aureus can also alter the apoptotic fate of the neutrophil and potentially skew other important signalling pathways for its own gain. Understanding these critical host-pathogen interactions could lead to the development of new host directed therapeutics for the treatment of S. aureus infection by removing its intracellular niche and restoring host bactericidal functions. This review discusses the current findings surrounding intracellular survival of S. aureus within neutrophils, the pathogenic role autophagy plays in this process and considers the therapeutic potential for targeting this immune evasion mechanism.
Collapse
Affiliation(s)
- Emilio G Vozza
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Michelle E Mulcahy
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Rachel M McLoughlin
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
19
|
Pérez C, Zúñiga T, Palavecino CE. Photodynamic therapy for treatment of Staphylococcus aureus infections. Photodiagnosis Photodyn Ther 2021; 34:102285. [PMID: 33836278 DOI: 10.1016/j.pdpdt.2021.102285] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/10/2021] [Accepted: 04/02/2021] [Indexed: 12/09/2022]
Abstract
BACKGROUND Staphylococcus aureus is a Gram-positive spherical bacterium that commonly causes various infections which can range from superficial to life-threatening. Hospital strains of S. aureus are often resistant to antibiotics, which has made their treatment difficult in recent decades. Other therapeutic alternatives have been postulated to overcome the drawbacks of antibiotic multi-resistance. Of these, photodynamic therapy (PDT) is a promising approach to address the notable shortage of new active antibiotics against multidrug-resistant S. aureus. PDT combines the use of a photosensitizer agent, light, and oxygen to eradicate pathogenic microorganisms. Through a systematic analysis of published results, this work aims to verify the usefulness of applying PDT in treating multidrug-resistant S.aureus infections. METHODS This review was based on a bibliographic search in various databases and the analysis of relevant publications. RESULTS There is currently a large body of evidence demonstrating the efficacy of photodynamic therapy in eliminating S.aureus strains. Both biofilm-producing strains, as well as multidrug-resistant strains. CONCLUSION We conclude that there is sufficient scientific evidence that PDT is a useful adjunct to traditional antibiotic therapy for treating S. aureus infections. Clinical application through appropriate trials should be introduced to further define optimal treatment protocols, safety and efficay.
Collapse
Affiliation(s)
- Camila Pérez
- Escuela de Tecnología Médica, Facultad de Ciencias de la Salud, Universidad Central de Chile, Chile.
| | - Tania Zúñiga
- Escuela de Tecnología Médica, Facultad de Ciencias de la Salud, Universidad Central de Chile, Chile.
| | - Christian Erick Palavecino
- Laboratorio de Microbiología Celular, Instituto de Investigación e Innovación en Salud, Facultad de Ciencias de la Salud, Universidad Central de Chile, Lord Cochrane 418, 8330546, Santiago, Chile.
| |
Collapse
|
20
|
Immune System Evasion Mechanisms in Staphylococcus aureus: Current Understanding. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2020. [DOI: 10.22207/jpam.14.4.01] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Staphylococcus aureus is a major human pathogen that may cause a wide range of infections and is a frequent cause of soft tissue and bloodstream infections. It is a successful pathogen due to its collective virulence factors and its ability to evade the host immune systems. The review aims to highlight how S. aureus destroys and damage the host cells and explains how immune cells can respond to this pathogen. This review may also provide new insights that may be useful for developing new strategy for combating MRSA and its emerging clones such as community-associated methicillin-resistant S. aureus (CA-MRSA).
Collapse
|
21
|
Algorri M, Wong-Beringer A. Differential effects of antibiotics on neutrophils exposed to lipoteichoic acid derived from Staphylococcus aureus. Ann Clin Microbiol Antimicrob 2020; 19:50. [PMID: 33143710 PMCID: PMC7641855 DOI: 10.1186/s12941-020-00392-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 10/20/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Persistent bacteremia occurs in at least 30% of patients with Staphylococcus aureus bloodstream infection (SAB) and may be attributable to a dysregulated host immune response. Neutrophils interact with a variety of S. aureus microbial factors, including lipoteichoic acid (LTA), to activate phagocytic function in a concentration-dependent manner. Antibiotics have been shown to exert both direct antimicrobial action as well as immunomodulatory effects. In this study, we compared the effects of different anti-staphylococcal antibiotics on LTA-mediated immune activation of neutrophils. METHODS Neutrophils obtained from healthy volunteers were exposed to two levels of LTA (1 and 10 μg/ml) with or without addition of antibiotics from different pharmacologic classes (vancomycin, daptomycin, ceftaroline). Neutrophil function was assessed by examining phagocytic response, activation (CD11b, CD62L expression), Toll-like receptor-2 expression, cell survival and apoptosis, and CXCL8 release. RESULTS Differential LTA-mediated antibiotic effects on neutrophil function were observed primarily at the high LTA exposure level. Ceftaroline in the presence of 10 μg/ml LTA had the most prominent effects on phagocytosis and CD11b and CD62L expression, with trends towards increased neutrophil survival and preservation of CXCL8 release when compared to daptomycin and vancomycin with the latter significantly dampening PMN CXCL8 release. CONCLUSIONS Select antimicrobial agents, such as ceftaroline, exert immunostimulatory effects on neutrophils exposed to S. aureus LTA, which when confirmed in vivo, could be leveraged for its dual immunomodulatory and antibacterial actions for the treatment of persistent SAB mediated by a dysregulated host response.
Collapse
Affiliation(s)
- Marquerita Algorri
- University of Southern California, School of Pharmacy, 1985 Zonal Avenue, Los Angeles, CA, 90089, USA
| | - Annie Wong-Beringer
- University of Southern California, School of Pharmacy, 1985 Zonal Avenue, Los Angeles, CA, 90089, USA.
| |
Collapse
|
22
|
Mrochen DM, Fernandes de Oliveira LM, Raafat D, Holtfreter S. Staphylococcus aureus Host Tropism and Its Implications for Murine Infection Models. Int J Mol Sci 2020; 21:E7061. [PMID: 32992784 PMCID: PMC7582387 DOI: 10.3390/ijms21197061] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/22/2020] [Accepted: 09/22/2020] [Indexed: 12/11/2022] Open
Abstract
Staphylococcus aureus (S. aureus) is a pathobiont of humans as well as a multitude of animal species. The high prevalence of multi-resistant and more virulent strains of S. aureus necessitates the development of new prevention and treatment strategies for S. aureus infection. Major advances towards understanding the pathogenesis of S. aureus diseases have been made using conventional mouse models, i.e., by infecting naïve laboratory mice with human-adapted S.aureus strains. However, the failure to transfer certain results obtained in these murine systems to humans highlights the limitations of such models. Indeed, numerous S. aureus vaccine candidates showed promising results in conventional mouse models but failed to offer protection in human clinical trials. These limitations arise not only from the widely discussed physiological differences between mice and humans, but also from the lack of attention that is paid to the specific interactions of S. aureus with its respective host. For instance, animal-derived S. aureus lineages show a high degree of host tropism and carry a repertoire of host-specific virulence and immune evasion factors. Mouse-adapted S.aureus strains, humanized mice, and microbiome-optimized mice are promising approaches to overcome these limitations and could improve transferability of animal experiments to human trials in the future.
Collapse
Affiliation(s)
- Daniel M. Mrochen
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse DZ 7, 17475 Greifswald, Germany; (L.M.F.d.O.); (D.R.); (S.H.)
| | - Liliane M. Fernandes de Oliveira
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse DZ 7, 17475 Greifswald, Germany; (L.M.F.d.O.); (D.R.); (S.H.)
| | - Dina Raafat
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse DZ 7, 17475 Greifswald, Germany; (L.M.F.d.O.); (D.R.); (S.H.)
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, 21521 Alexandria, Egypt
| | - Silva Holtfreter
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Strasse DZ 7, 17475 Greifswald, Germany; (L.M.F.d.O.); (D.R.); (S.H.)
| |
Collapse
|
23
|
Alhede M, Lorenz M, Fritz BG, Jensen PØ, Ring HC, Bay L, Bjarnsholt T. Bacterial aggregate size determines phagocytosis efficiency of polymorphonuclear leukocytes. Med Microbiol Immunol 2020; 209:669-680. [PMID: 32880037 PMCID: PMC7568703 DOI: 10.1007/s00430-020-00691-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 08/19/2020] [Indexed: 02/06/2023]
Abstract
The ability of bacteria to aggregate and form biofilms impairs phagocytosis by polymorphonuclear leukocytes (PMNs). The aim of this study was to examine if the size of aggregates is critical for successful phagocytosis and how bacterial biofilms evade phagocytosis. We investigated the live interaction between PMNs and Pseudomonas aeruginosa, Staphylococcus aureus, Escherichia coli and Staphylococcus epidermidis using confocal scanning laser microscopy. Aggregate size significantly affected phagocytosis outcome and larger aggregates were less likely to be phagocytized. Aggregates of S. epidermidis were also less likely to be phagocytized than equally-sized aggregates of the other three species. We found that only aggregates of approx. 5 μm diameter or smaller were consistently phagocytosed. We demonstrate that planktonic and aggregated cells of all four species significantly reduced the viability of PMNs after 4 h of incubation. Our results indicate that larger bacterial aggregates are less likely to be phagocytosed by PMNs and we propose that, if the aggregates become too large, circulating PMNs may not be able to phagocytose them quickly enough, which may lead to chronic infection.
Collapse
Affiliation(s)
- Maria Alhede
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Melanie Lorenz
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Blaine Gabriel Fritz
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Peter Østrup Jensen
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark.,Department of Clinical Microbiology, Rigshospitalet, Afsnit 9301, Juliane Maries Vej 22, DK-2100, Copenhagen Ø, Denmark.,Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, 2100, Copenhagen, Denmark
| | - Hans Christian Ring
- Department of Dermatology, Bispebjerg Hospital, Nielsine Nielsens Vej 9, København, NV, Denmark
| | - Lene Bay
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Thomas Bjarnsholt
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark. .,Department of Clinical Microbiology, Rigshospitalet, Afsnit 9301, Juliane Maries Vej 22, DK-2100, Copenhagen Ø, Denmark.
| |
Collapse
|
24
|
Sultana S, Bishayi B. Etoposide-mediated depletion of peripheral blood monocytes post s.aureus infection attenuates septic arthritis by modulating macrophage-derived factors responsible for inflammatory destruction. Immunol Lett 2020; 220:51-62. [PMID: 32032616 DOI: 10.1016/j.imlet.2020.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/27/2020] [Accepted: 02/01/2020] [Indexed: 01/16/2023]
Abstract
S.aureus induced septic arthritis remains a serious medical concern due to its rapidly progressive disease profile. The multidrug resistant nature of S.aureus demands the development of new strategies for the treatment of S.aureus arthritis. Since monocyte/macrophage population has been recognized as an important axis in joint inflammation and destruction, selective depletion of peripheral blood monocytes might influence the outcome and progression of the disease. Therefore, in this study we have put forward the concept of monocyte depletion by using etoposide, a drug that selectively depletes the monocyte/macrophage population. Mice were inoculated with live S.aureus for the development of septic arthritis. Post S.aureus infection, etoposide was subcutaneously injected. The severity of arthritis was found to be significantly low in the etoposide treated mice throughout the course. Arthritis index, histopathological analysis and TRAP staining images confirmed effectiveness of etoposide treatment in regulating inflammation and bone cartilage destruction. Lower levels of inflammatory cytokines, ROS, MMP-2, RANKL, OPN and plasmin reflected less severe arthritic destruction after etoposide treatment in arthritic mice. The bacterial load was not increased after etoposide treatment. Together, the presented data suggested that monocyte depletion by etoposide might represent an alternative therapeutic strategy for the treatment of S.aureus arthritis.
Collapse
Affiliation(s)
- Sahin Sultana
- Department of Physiology, Immunology and Microbiology laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta, 700009, West Bengal, India
| | - Biswadev Bishayi
- Department of Physiology, Immunology and Microbiology laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta, 700009, West Bengal, India.
| |
Collapse
|
25
|
In Staphylococcus aureus, the Particulate State of the Cell Envelope Is Required for the Efficient Induction of Host Defense Responses. Infect Immun 2019; 87:IAI.00674-19. [PMID: 31548327 DOI: 10.1128/iai.00674-19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 09/16/2019] [Indexed: 02/07/2023] Open
Abstract
Upon microbial infection, host immune cells recognize bacterial cell envelope components through cognate receptors. Although bacterial cell envelope components function as innate immune molecules, the role of the physical state of the bacterial cell envelope (i.e., particulate versus soluble) in host immune activation has not been clearly defined. Here, using two different forms of the staphylococcal cell envelope of Staphylococcus aureus RN4220 and USA300 LAC strains, we provide biochemical and immunological evidence that the particulate state is required for the effective activation of host innate immune responses. In a murine model of peritoneal infection, the particulate form of the staphylococcal cell envelope (PCE) induced the production of chemokine (C-X-C motif) ligand 1 (CXCL1) and CC chemokine ligand 2 (CCL2), the chemotactic cytokines for neutrophils and monocytes, respectively, resulting in a strong influx of the phagocytes into the peritoneal cavity. In contrast, compared with PCE, the soluble form of cell envelope (SCE), which was derived from PCE by treatment with cell wall-hydrolyzing enzymes, showed minimal activity. PCE also induced the secretion of calprotectin (myeloid-related protein 8/14 [MRP8/14] complex), a phagocyte-derived antimicrobial protein, into the peritoneal cavity at a much higher level than did SCE. The injected PCE particles were phagocytosed by the infiltrated neutrophils and monocytes and then delivered to mediastinal draining lymph nodes. More importantly, intraperitoneally (i.p.) injected PCE efficiently protected mice from S. aureus infection, which was abolished by the depletion of either monocytes/macrophages or neutrophils. This study demonstrated that the physical state of bacterial cells is a critical factor for efficient host immune activation and the protection of hosts from staphylococcal infections.
Collapse
|
26
|
Maslikov SN, Bely DD, Samoiliuk VV, Vakulik VV, Spitsyna TL. Pathogenetic role of Staphylococcus aureus in purulent keratoconjunctivitis in cats. REGULATORY MECHANISMS IN BIOSYSTEMS 2019. [DOI: 10.15421/021974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The research was carried out in the Department of Surgery and Obstetrics of Agricultural Animals of Dnipro State Agrarian and Economic University on clinically healthy outbred cats of different ages with purulent keratoconjunctivitis. Hematological, biochemical and immunological parameters were determined in the animals, and microbiological and virological research was conducted on them. According to the data obtained, more than half of cases of ophthalmopathology in cats were conjunctivitis and keratoconjunctivitis, and they were more often registered in the cold season. The main causes of eye diseases in the cats were mechanical injuries, coccal and chlamydial infection, allergy and development of disease against the background of primary lesions of the ears and paranasal sinuses. Among the detected microorganisms, the vast majority (81.9%) were staphylococci, including S. albus, S. aureus and S. epidermidis. All types of microorganisms except O-forms of Bacillus sp. exhibited high and medium sensitivity to antibiotics. Immunoblotting revealed polypeptides that responded to specific S. aureus antigens in samples of conjunctiva, cornea, intraocular fluid, and blood of cats suffering from purulent (staphylococcal) keratoconjunctivitis. The highest antigen concentration was detected in the cornea and conjunctiva. In the absence of expressive shifts of the investigated hematological and biochemical parameters, the dynamics of immunological markers were shown by a significant increase in the content of immunoglobulins and circulating immune complexes, as well as by a distinct activation of the complement system by the classical route. The results showed a clear gradual activation of phagocytosis, namely: the number of phagocytic neutrophils increased, reaching its maximum value by the seventh day of development of purulent keratoconjunctivitis. The phagocytic index in the first three days of observation tended to decrease, and by the seventh day it had already significantly exceeded the initial value. Despite the ambiguous dynamics of the phagocytic index, in the course of the development of the inflammatory process in the blood of sick cats, we observed a clear increase in the index of complete phagocytosis. Thus, the development of purulent keratoconjunctivitis in cats occurs against the background of clear cellular and humoral responses to the infectious agent.
Collapse
|
27
|
Arciola CR, Campoccia D, Montanaro L. Implant infections: adhesion, biofilm formation and immune evasion. Nat Rev Microbiol 2019; 16:397-409. [PMID: 29720707 DOI: 10.1038/s41579-018-0019-y] [Citation(s) in RCA: 1091] [Impact Index Per Article: 218.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Medical device-associated infections account for a large proportion of hospital-acquired infections. A variety of opportunistic pathogens can cause implant infections, depending on the type of the implant and on the anatomical site of implantation. The success of these versatile pathogens depends on rapid adhesion to virtually all biomaterial surfaces and survival in the hostile host environment. Biofilm formation on implant surfaces shelters the bacteria and encourages persistence of infection. Furthermore, implant-infecting bacteria can elude innate and adaptive host defences as well as biocides and antibiotic chemotherapies. In this Review, we explore the fundamental pathogenic mechanisms underlying implant infections, highlighting orthopaedic implants and Staphylococcus aureus as a prime example, and discuss innovative targets for preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Carla Renata Arciola
- Research Unit on Implant Infections, Rizzoli Orthopaedic Institute, Bologna, Italy. .,Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.
| | - Davide Campoccia
- Research Unit on Implant Infections, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Lucio Montanaro
- Research Unit on Implant Infections, Rizzoli Orthopaedic Institute, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| |
Collapse
|
28
|
Josse J, Valour F, Maali Y, Diot A, Batailler C, Ferry T, Laurent F. Interaction Between Staphylococcal Biofilm and Bone: How Does the Presence of Biofilm Promote Prosthesis Loosening? Front Microbiol 2019; 10:1602. [PMID: 31379772 PMCID: PMC6653651 DOI: 10.3389/fmicb.2019.01602] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 06/26/2019] [Indexed: 12/19/2022] Open
Abstract
With the aging of population, the number of indications for total joint replacement is continuously increasing. However, prosthesis loosening can happen and is related to two major mechanisms: (1) aseptic loosening due to prosthesis micromotion and/or corrosion and release of wear particles from the different components of the implanted material and (2) septic loosening due to chronic prosthetic joint infection (PJI). The “aseptic” character of prosthesis loosening has been challenged over the years, especially considering that bacteria can persist in biofilms and be overlooked during diagnosis. Histological studies on periprosthetic tissue samples reported that macrophages are the principle cells associated with aseptic loosening due to wear debris. They produce cytokines and favor an inflammatory environment that induces formation and activation of osteoclasts, leading to bone resorption and periprosthetic osteolysis. In PJIs, the presence of infiltrates of polymorphonuclear neutrophils is a major criterion for histological diagnosis. Neutrophils are colocalized with osteoclasts and zones of osteolysis. A similar inflammatory environment also develops, leading to bone resorption through osteoclasts. Staphylococcus aureus, Staphylococcus epidermidis, and Staphylococcus lugdunensis are the main staphylococci observed in PJIs. They share the common feature to form biofilm. For S. aureus and S. epidermidis, the interaction between biofilm and immunes cells (macrophages and polymorphonuclear neutrophils) differs regarding the species. Indeed, the composition of extracellular matrix of biofilm seems to impact the interaction with immune cells. Recent papers also reported the major role of myeloid-derived suppressor cells in biofilm-associated PJIs with S. aureus. These cells prevent lymphocyte infiltration and facilitate biofilm persistence. Moreover, the role of T lymphocytes is still unclear and potentially underestimates. In this review, after introducing the cellular mechanism of aseptic and septic loosening, we will focus on the interrelationships between staphylococcal biofilm, immune cells, and bone cells.
Collapse
Affiliation(s)
- Jérôme Josse
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France
| | - Florent Valour
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Service de Chirurgie Orthopédique, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Yousef Maali
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Alan Diot
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Cécile Batailler
- Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Service de Maladies Infectieuses, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Tristan Ferry
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Service de Chirurgie Orthopédique, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Frédéric Laurent
- CIRI - Centre International de Recherche en Infectiologie, Inserm U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France.,Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Centre Interrégional de Référence des Infections Ostéo-articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Laboratoire de Bactériologie, Institut des Agents Infectieux, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
29
|
Mostafa E, Fayed MAA, Radwan RA, Bakr RO. Centaurea pumilio L. extract and nanoparticles: A candidate for healthy skin. Colloids Surf B Biointerfaces 2019; 182:110350. [PMID: 31326622 DOI: 10.1016/j.colsurfb.2019.110350] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/14/2019] [Accepted: 07/06/2019] [Indexed: 01/09/2023]
Abstract
Centaurea pumilio was the subject of phytochemical and biological studies, and its extract was used in the green synthesis of silver nanoparticles (AgNPs). Liquid chromatography/electrospray ionization mass spectrometry allowed the tentative identification of twenty-nine phytoconstituents of C. pumilio methanolic extract (CME), while column chromatography led to the identification of eight phenolic compounds. The neutral red uptake method showed the safety of CME and AgNPs on skin cells (HaCaT cell lines), while their high antioxidant potentials were demonstrated based on their oxygen radical absorbance capacity, and these results were confirmed in vivo. Additionally, CME and AgNPs had promising abilities to retard the ageing process and combat dark spots by potently inhibiting collagenase, elastase and tyrosinase, in addition to antimicrobial activity against skin infection-causing strains, especially Staphylococcus aureus, which was further confirmed by the significant phagocytic activity of neutrophils via engulfment. This study presents C. pumilio as a candidate for healthy skin.
Collapse
Affiliation(s)
- Eman Mostafa
- Department of Pharmacognosy, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), 11787, Giza, Egypt
| | - Marwa A A Fayed
- Department of Pharmacognosy, Faculty of Pharmacy, University of Sadat City, Egypt
| | - Rasha A Radwan
- Biochemistry and Biotechnology Department, Faculty of Pharmacy & Drug Technology, Heliopolis University, Cairo, Egypt
| | - Riham O Bakr
- Department of Pharmacognosy, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), 11787, Giza, Egypt.
| |
Collapse
|
30
|
Sward EW, Fones EM, Spaan RR, Pallister KB, Haller BL, Guerra FE, Zurek OW, Nygaard TK, Voyich JM. Staphylococcus aureus SaeR/S-Regulated Factors Decrease Monocyte-Derived Tumor Necrosis Factor-α to Reduce Neutrophil Bactericidal Activity. J Infect Dis 2019; 217:943-952. [PMID: 29272502 DOI: 10.1093/infdis/jix652] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/17/2017] [Indexed: 12/11/2022] Open
Abstract
Background The ability of Staphylococcus aureus to evade killing by human neutrophils significantly contributes to disease progression. In this study, we characterize an influential role for the S. aureus SaeR/S 2-component gene regulatory system in suppressing monocyte production of tumor necrosis factor alpha (TNF-α) to subsequently influence human neutrophil priming. Methods Using flow cytometry and TNF-α specific enzyme-linked immunosorbent assays we identify the primary cellular source of TNF-α in human blood and in purified peripheral blood mononuclear cells (PBMCs) during interaction with USA300 and an isogenic saeR/S deletion mutant (USA300∆saeR/S). Assays with conditioned media from USA300 and USA300∆saeR/S exposed PBMCs were used to investigate priming on neutrophil bactericidal activity. Results TNF-α production from monocytes was significantly reduced following challenge with USA300 compared to USA300∆saeR/S. We observed that priming of neutrophils using conditioned medium from peripheral blood mononuclear cells stimulated with USA300∆saeR/S significantly increased neutrophil bactericidal activity against USA300 relative to unprimed neutrophils and neutrophils primed with USA300 conditioned medium. The increased neutrophil bactericidal activity was associated with enhanced reactive oxygen species production that was significantly influenced by elevated TNF-α concentrations. Conclusions Our findings identify an immune evasion strategy used by S. aureus to impede neutrophil priming and subsequent bactericidal activity.
Collapse
Affiliation(s)
- Eli W Sward
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Elizabeth M Fones
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Russel R Spaan
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Kyler B Pallister
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Brandon L Haller
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Fermin E Guerra
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Oliwia W Zurek
- Infectious Disease Department, Genentech Inc, South San Francisco, California
| | - Tyler K Nygaard
- Department of Microbiology and Immunology, Montana State University, Bozeman
| | - Jovanka M Voyich
- Department of Microbiology and Immunology, Montana State University, Bozeman
| |
Collapse
|
31
|
Elshina E, Allen ER, Flaxman A, van Diemen PM, Milicic A, Rollier CS, Yamaguchi Y, Wyllie DH. Vaccination with the Staphylococcus aureus secreted proteins EapH1 and EapH2 impacts both S. aureus carriage and invasive disease. Vaccine 2018; 37:502-509. [PMID: 30502067 DOI: 10.1016/j.vaccine.2018.11.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 11/02/2018] [Accepted: 11/13/2018] [Indexed: 12/15/2022]
Abstract
INTRODUCTION There is a need for an efficacious vaccine reducing infections due to Staphylococcus aureus, a common cause of community and hospital infection. Infecting organisms originate from S. aureus populations colonising the nares and bowel. Antimicrobials are widely used to transiently reduce S. aureus colonisation prior to surgery, a practice which is selecting for resistant S. aureus isolates. S. aureus secretes multiple proteins, including the protease inhibitors extracellular adhesion protein homologue 1 and 2 (EapH1 and EapH2). METHODS Mice were vaccinated intramuscularly or intranasally with Adenovirus serotype 5 and Modified Vaccinia Ankara viral vectors expressing EapH1 and EapH2 proteins, or with control viruses. Using murine S. aureus colonisation models, we monitored S. aureus colonisation by sequential stool sampling. Monitoring of S. aureus invasive disease after intravenous challenge was performed using bacterial load and abscess numbers in the kidney. RESULTS Intramuscular vaccination with Adenovirus serotype 5 and Modified Vaccinia Ankara viral vectors expressing EapH1 and EapH2 proteins significantly reduces bacterial recovery in the murine renal abscess model of infection, but the magnitude of the effect is small. A single intranasal vaccination with an adenoviral vaccine expressing these proteins reduced S. aureus gastrointestinal (GI) tract colonisation. CONCLUSION Vaccination against EapH1 / EapH2 proteins may offer an antibiotic independent way to reduce S. aureus colonisation, as well as contributing to protection against S. aureus invasive disease.
Collapse
Affiliation(s)
- Elizaveta Elshina
- Jenner Institute, Centre for Cellular and Molecular Physiology, University of Oxford, United Kingdom
| | - Elizabeth R Allen
- Jenner Institute, Centre for Cellular and Molecular Physiology, University of Oxford, United Kingdom
| | - Amy Flaxman
- Jenner Institute, Centre for Cellular and Molecular Physiology, University of Oxford, United Kingdom
| | - Pauline M van Diemen
- Jenner Institute, Centre for Cellular and Molecular Physiology, University of Oxford, United Kingdom
| | - Anita Milicic
- Jenner Institute, Centre for Cellular and Molecular Physiology, University of Oxford, United Kingdom
| | - Christine S Rollier
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and NIHR Oxford Biomedical Research Centre, Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, United Kingdom
| | - Yuko Yamaguchi
- Jenner Institute, Centre for Cellular and Molecular Physiology, University of Oxford, United Kingdom
| | - David H Wyllie
- Jenner Institute, Centre for Cellular and Molecular Physiology, University of Oxford, United Kingdom.
| |
Collapse
|
32
|
Heterogeneity of Ly6G + Ly6C + Myeloid-Derived Suppressor Cell Infiltrates during Staphylococcus aureus Biofilm Infection. Infect Immun 2018; 86:IAI.00684-18. [PMID: 30249747 DOI: 10.1128/iai.00684-18] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 09/20/2018] [Indexed: 01/11/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature monocytes and granulocytes. While neutrophils (polymorphonuclear leukocytes [PMNs]) are classically identified as highly differentiated cells specialized for antimicrobial defense, our laboratory has reported minor contributions of PMNs to the immune response during Staphylococcus aureus biofilm infection. However, these two cell types can be difficult to differentiate because of shared surface marker expression. Here we describe a more refined approach to distinguish MDSCs from PMNs utilizing the integrin receptor CD11b combined with conventional Ly6G and Ly6C expression. This approach separated the Ly6G+ Ly6C+ population that we previously identified in a mouse model of S. aureus orthopedic implant infection into two subsets, namely, CD11bhigh Ly6G+ Ly6C+ MDSCs and CD11blow Ly6G+ Ly6C+ PMNs, which was confirmed by characteristic nuclear morphology using cytospins. CD11bhigh Ly6G+ Ly6C+ MDSCs suppressed T cell proliferation throughout the 28-day infection period, whereas CD11blow Ly6G+ Ly6C+ PMNs had no effect early (day 3 postinfection), although this population acquired suppressive activity at later stages of biofilm development. To further highlight the distinctions between biofilm-associated MDSCs and PMNs versus monocytes, transcriptional profiles were compared by transcriptome sequencing (RNA-Seq). A total of 6,466 genes were significantly differentially expressed in MDSCs versus monocytes, whereas only 297 genes were significantly different between MDSCs and PMNs. A number of genes implicated in cell cycle regulation were identified, and in vivo ethynyldeoxyuridine (EdU) labeling revealed that approximately 50% of MDSCs proliferated locally at the site of S. aureus biofilm infection. Based on their similar transcriptomic profiles to those of PMNs, biofilm-associated MDSCs are of a granulocytic lineage and can be classified as granulocytic MDSCs (G-MDSCs).
Collapse
|
33
|
Perez K, Patel R. Survival of Staphylococcus epidermidis in Fibroblasts and Osteoblasts. Infect Immun 2018; 86:e00237-18. [PMID: 30061380 PMCID: PMC6204734 DOI: 10.1128/iai.00237-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/20/2018] [Indexed: 12/26/2022] Open
Abstract
Staphylococcus epidermidis is a leading cause of infections associated with indwelling medical devices, including prosthetic joint infection. While biofilm formation is assumed to be the main mechanism underlying the chronic infections S. epidermidis causes, we hypothesized that S. epidermidis also evades immune killing, contributing to its pathogenesis. Here, we show that prosthetic joint-associated S. epidermidis isolates can persist intracellularly within human fibroblasts and inside human and mouse osteoblasts. We also show that the intracellularly persisting bacteria reside primarily within acidic phagolysosomes and that over the course of infection, small-colony variants are selected for. Moreover, upon eukaryotic cell death, these bacteria, which can outlive their host, can escape into the extracellular environment, providing them an opportunity to form biofilms on implant surfaces at delayed time points in implant-associated infection. In summary, the acidic phagolysosomes of fibroblasts and osteoblasts serve as reservoirs for chronic or delayed S. epidermidis infection.
Collapse
Affiliation(s)
- Kimberly Perez
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
34
|
Krishna A, Holden MTG, Peacock SJ, Edwards AM, Wigneshweraraj S. Naturally occurring polymorphisms in the virulence regulator Rsp modulate Staphylococcus aureus survival in blood and antibiotic susceptibility. MICROBIOLOGY (READING, ENGLAND) 2018; 164:1189-1195. [PMID: 30028663 PMCID: PMC6230762 DOI: 10.1099/mic.0.000695] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/03/2018] [Indexed: 01/20/2023]
Abstract
Nasal colonization by the pathogen Staphylococcus aureus is a risk factor for subsequent infection. Loss of function mutations in the gene encoding the virulence regulator Rsp are associated with the transition of S. aureus from a colonizing isolate to one that causes bacteraemia. Here, we report the identification of several novel activity-altering mutations in rsp detected in clinical isolates, including for the first time, mutations that enhance agr operon activity. We assessed how these mutations affected infection-relevant phenotypes and found loss and enhancement of function mutations to have contrasting effects on S. aureus survival in blood and antibiotic susceptibility. These findings add to the growing body of evidence that suggests S. aureus 'trades off' virulence for the acquisition of traits that benefit survival in the host, and indicates that infection severity and treatment options can be significantly affected by mutations in the virulence regulator rsp.
Collapse
Affiliation(s)
- Aishwarya Krishna
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Matthew T. G. Holden
- Wellcome Trust Sanger Institute, Hinxton, UK
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Sharon J. Peacock
- Wellcome Trust Sanger Institute, Hinxton, UK
- London School of Hygiene and Tropical Medicine, London, UK
| | - Andrew M. Edwards
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | | |
Collapse
|
35
|
MntC-Dependent Manganese Transport Is Essential for Staphylococcus aureus Oxidative Stress Resistance and Virulence. mSphere 2018; 3:3/4/e00336-18. [PMID: 30021878 PMCID: PMC6052334 DOI: 10.1128/msphere.00336-18] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Work outlined in this report demonstrated that MntC-dependent manganese transport is required for S. aureus virulence. These study results support the model that MntC-specific antibodies elicited by a vaccine have the potential to disrupt S. aureus manganese transport and thus abrogate to its virulence. Staphylococcus aureus is a human pathogen that has developed several approaches to evade the immune system, including a strategy to resist oxidative killing by phagocytes. This resistance is mediated by production of superoxide dismutase (SOD) enzymes which use manganese as a cofactor. S. aureus encodes two manganese ion transporters, MntABC and MntH, and a possible Nramp family manganese transporter, exemplified by S. aureus N315 SA1432. Their relative contributions to manganese transport have not been well defined in clinically relevant isolates. For this purpose, insertional inactivation mutations were introduced into mntC, mntH, and SA1432 individually and in combination. mntC was necessary for full resistance to methyl viologen, a compound that generates intracellular free radicals. In contrast, strains with an intact mntH gene had a minimal increase in resistance that was revealed only in mntC strains, and no change was observed upon mutation of SA1432 in strains lacking both mntC and mntH. Similarly, MntC alone was required for high cellular SOD activity. In addition, mntC strains were attenuated in a murine sepsis model. To further link these observations to manganese transport, an S. aureus MntC protein lacking manganese binding activity was designed, expressed, and purified. While circular dichroism experiments demonstrated that the secondary and tertiary structures of this protein were unaltered, a defect in manganese binding was confirmed by isothermal titration calorimetry. Unlike complementation with wild-type mntC, introduction of the manganese-binding defective allele into the chromosome of an mntC strain did not restore resistance to oxidative stress or virulence. Collectively, these results underscore the importance of MntC-dependent manganese transport in S. aureus oxidative stress resistance and virulence. IMPORTANCE Work outlined in this report demonstrated that MntC-dependent manganese transport is required for S. aureus virulence. These study results support the model that MntC-specific antibodies elicited by a vaccine have the potential to disrupt S. aureus manganese transport and thus abrogate to its virulence.
Collapse
|
36
|
Tuffs SW, Haeryfar SMM, McCormick JK. Manipulation of Innate and Adaptive Immunity by Staphylococcal Superantigens. Pathogens 2018; 7:pathogens7020053. [PMID: 29843476 PMCID: PMC6027230 DOI: 10.3390/pathogens7020053] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 12/15/2022] Open
Abstract
Staphylococcal superantigens (SAgs) constitute a family of potent exotoxins secreted by Staphylococcus aureus and other select staphylococcal species. SAgs function to cross-link major histocompatibility complex (MHC) class II molecules with T cell receptors (TCRs) to stimulate the uncontrolled activation of T lymphocytes, potentially leading to severe human illnesses such as toxic shock syndrome. The ubiquity of SAgs in clinical S. aureus isolates suggests that they likely make an important contribution to the evolutionary fitness of S. aureus. Although the apparent redundancy of SAgs in S. aureus has not been explained, the high level of sequence diversity within this toxin family may allow for SAgs to recognize an assorted range of TCR and MHC class II molecules, as well as aid in the avoidance of humoral immunity. Herein, we outline the major diseases associated with the staphylococcal SAgs and how a dysregulated immune system may contribute to pathology. We then highlight recent research that considers the importance of SAgs in the pathogenesis of S. aureus infections, demonstrating that SAgs are more than simply an immunological diversion. We suggest that SAgs can act as targeted modulators that drive the immune response away from an effective response, and thus aid in S. aureus persistence.
Collapse
Affiliation(s)
- Stephen W Tuffs
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada.
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada.
- Division of Clinical Immunology and Allergy, Department of Medicine, Western University, London, ON N6A 3K7, Canada.
- Centre for Human Immunology, Western University, London, ON N6A 3K7, Canada.
- Lawson Health Research Institute, London, ON N6C 2R5, Canada.
| | - John K McCormick
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada.
- Centre for Human Immunology, Western University, London, ON N6A 3K7, Canada.
- Lawson Health Research Institute, London, ON N6C 2R5, Canada.
| |
Collapse
|
37
|
Interactions of staphyloxanthin and enterobactin with myeloperoxidase and reactive chlorine species. Arch Biochem Biophys 2018; 646:80-89. [PMID: 29614256 DOI: 10.1016/j.abb.2018.03.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/29/2018] [Accepted: 03/30/2018] [Indexed: 12/19/2022]
Abstract
When neutrophils engulf bacteria, myeloperoxidase converts hydrogen peroxide to hypochlorous acid, which is toxic to all micro-organisms. It has been suggested that some pathogens have virulence factors that target myeloperoxidase to dampen the oxidative reactions of neutrophils. These virulence factors include staphyloxanthin, the golden pigment of Staphylococcus aureus, and enterobactin - a siderophore released by gram-negative bacteria. We investigated the potential of staphyloxanthin and enterobactin to shield bacteria from hypochlorous acid and related chloramines. Clinical strains of S. aureus with high levels of staphyloxanthin and related carotenoids were in general more resistant to low doses of hypochlorous acid than non-pigmented bacteria. But some non-pigmented strains were also resistant to the oxidant. Doses of reactive chlorine species that killed bacteria also bleached their carotenoids. Hypochlorous acid, NH2Cl, and NHCl2 bleached purified staphyloxanthin. When S. aureus were phagocytosed by neutrophils there was no discernible loss of staphyloxanthin. These data suggest that staphyloxanthin is capable of protecting bacteria from low doses of reactive chlorine species formed inside phagosomes. Enterobactin was not an inhibitor of myeloperoxidase. We conclude that staphyloxanthin may protect some bacterial strains against oxidative killing by neutrophils, but enterobactin will not inhibit the production of hypochlorous acid.
Collapse
|
38
|
Zhou Y, Niu C, Ma B, Xue X, Li Z, Chen Z, Li F, Zhou S, Luo X, Hou Z. Inhibiting PSMα-induced neutrophil necroptosis protects mice with MRSA pneumonia by blocking the agr system. Cell Death Dis 2018; 9:362. [PMID: 29500427 PMCID: PMC5834619 DOI: 10.1038/s41419-018-0398-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 02/06/2018] [Accepted: 02/12/2018] [Indexed: 12/11/2022]
Abstract
Given its high resistance, enhanced virulence, and high transmissibility, community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) pneumonia is highly associated with high morbidity and mortality. Anti-virulence therapy is a promising strategy that bypasses the evolutionary pressure on the bacterium to develop resistance. RNAIII-inhibiting peptide (RIP), as an accessory gene regulator (agr)-specific inhibitor, significantly restricts the virulence of S. aureus and protects infected mice from death by blocking the agr quorum sensing system. The protective effects of RIP on the neutropenic mice completely disappeared in a neutrophil-deleted mouse infection model, but not in the macrophage-deleted mice. This result confirmed that the in vivo antibacterial activity of RIP is highly associated with neutrophil function. Phenol-soluble modulins (PSMs), as major leukocyte lysis toxins of CA-MRSA, are directly regulated by the agr system. In this experiment, PSMα1, 2, and 3 significantly induced neutrophil necroptosis by activating mixed lineage kinase-like protein (MLKL) phosphorylation and increasing lactate dehydrogenase release. The S. aureus supernatants harvested from the agr or psmα mutant strains both decreased the phosphorylation level of MLKL and cell lysis. PSMα1-mediated neutrophil lysis was significantly inhibited by necrosulfonamide, necrostatin-1, TNFα antibody, and WRW4. These results showed PSMα1 induced necroptosis depends on formylpeptide receptor 2 (FPR2)-mediated autocrine TNFα. Moreover, the neutrophil necroptosis induced by S. aureus was significantly suppressed and pneumonia was effectively prevented by the blockage of agrA and psmα expression levels. These findings indicate that PSMα-induced necroptosis is a major cause of lung pathology in S. aureus pneumonia and suggest that interfering with the agr quorum sensing signaling pathway is a potential therapeutic strategy.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China.,Department of Pharmacology, Xi'an Medical University, Xi'an, China
| | - Chao Niu
- Henan Eye Institute, Henan Provincial People's Hospital, Zhengzhou, China
| | - Bo Ma
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Xiaoyan Xue
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Zhi Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Zhou Chen
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Fen Li
- Department of Pharmacology, Xi'an Medical University, Xi'an, China
| | - Shan Zhou
- Department of Clinical Laboratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaoxing Luo
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China.
| | - Zheng Hou
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
39
|
Varshney AK, Kuzmicheva GA, Lin J, Sunley KM, Bowling RA, Kwan TY, Mays HR, Rambhadran A, Zhang Y, Martin RL, Cavalier MC, Simard J, Shivaswamy S. A natural human monoclonal antibody targeting Staphylococcus Protein A protects against Staphylococcus aureus bacteremia. PLoS One 2018; 13:e0190537. [PMID: 29364906 PMCID: PMC5783355 DOI: 10.1371/journal.pone.0190537] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 12/14/2017] [Indexed: 11/18/2022] Open
Abstract
Staphylococcus aureus can cause devastating and life-threatening infections. With the increase in multidrug resistant strains, novel therapies are needed. Limited success with active and passive immunization strategies have been attributed to S. aureus immune evasion. Here, we report on a monoclonal antibody, 514G3, that circumvents a key S. aureus evasion mechanism by targeting the cell wall moiety Protein A (SpA). SpA tightly binds most subclasses of immunoglobulins via their Fc region, neutralizing effector function. The organism can thus shield itself with a protective coat of serum antibodies and render humoral immunity ineffective. The present antibody reactivity was derived from an individual with natural anti-SpA antibody titers. The monoclonal antibody is of an IgG3 subclass, which differs critically from other immunoglobulin subclasses since its Fc is not bound by SpA. Moreover, it targets a unique epitope on SpA that allows it to bind in the presence of serum antibodies. Consequently, the antibody opsonizes S. aureus and maintains effector function to enable natural immune mediated clearance. The data presented here provide evidence that 514G3 antibody is able to successfully rescue mice from S. aureus mediated bacteremia.
Collapse
Affiliation(s)
| | | | - Jian Lin
- XBiotech USA Inc., Austin, Texas, United States of America
| | | | | | - Tzu-Yu Kwan
- XBiotech USA Inc., Austin, Texas, United States of America
| | | | - Anu Rambhadran
- XBiotech USA Inc., Austin, Texas, United States of America
| | - Yanfeng Zhang
- XBiotech USA Inc., Austin, Texas, United States of America
| | | | | | - John Simard
- XBiotech USA Inc., Austin, Texas, United States of America
| | | |
Collapse
|
40
|
Kane TL, Carothers KE, Lee SW. Virulence Factor Targeting of the Bacterial Pathogen Staphylococcus aureus for Vaccine and Therapeutics. Curr Drug Targets 2018; 19:111-127. [PMID: 27894236 PMCID: PMC5957279 DOI: 10.2174/1389450117666161128123536] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/12/2016] [Accepted: 10/27/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND Staphylococcus aureus is a major bacterial pathogen capable of causing a range of infections in humans from gastrointestinal disease, skin and soft tissue infections, to severe outcomes such as sepsis. Staphylococcal infections in humans can be frequent and recurring, with treatments becoming less effective due to the growing persistence of antibiotic resistant S. aureus strains. Due to the prevalence of antibiotic resistance, and the current limitations on antibiotic development, an active and highly promising avenue of research has been to develop strategies to specifically inhibit the activity of virulence factors produced S. aureus as an alternative means to treat disease. OBJECTIVE In this review we specifically highlight several major virulence factors produced by S. aureus for which recent advances in antivirulence approaches may hold promise as an alternative means to treating diseases caused by this pathogen. Strategies to inhibit virulence factors can range from small molecule inhibitors, to antibodies, to mutant and toxoid forms of the virulence proteins. CONCLUSION The major prevalence of antibiotic resistant strains of S. aureus combined with the lack of new antibiotic discoveries highlight the need for vigorous research into alternative strategies to combat diseases caused by this highly successful pathogen. Current efforts to develop specific antivirulence strategies, vaccine approaches, and alternative therapies for treating severe disease caused by S. aureus have the potential to stem the tide against the limitations that we face in the post-antibiotic era.
Collapse
Affiliation(s)
- Trevor L. Kane
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Katelyn E. Carothers
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Shaun W. Lee
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
41
|
Lewis ML, Surewaard BGJ. Neutrophil evasion strategies by Streptococcus pneumoniae and Staphylococcus aureus. Cell Tissue Res 2017; 371:489-503. [PMID: 29204747 DOI: 10.1007/s00441-017-2737-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/06/2017] [Indexed: 02/05/2023]
Abstract
Humans are well equipped to defend themselves against bacteria. The innate immune system employs diverse mechanisms to recognize, control and initiate a response that can destroy millions of different microbes. Microbes that evade the sophisticated innate immune system are able to escape detection and could become pathogens. The pathogens Streptococcus pneumoniae and Staphylococcus aureus are particularly successful due to the development of a wide variety of virulence strategies for bacterial pathogenesis and they invest significant efforts towards mechanisms that allow for neutrophil evasion. Neutrophils are a primary cellular defense and can rapidly kill invading microbes, which is an indispensable function for maintaining host health. This review compares the key features of Streptococcus pneumoniae and Staphylococcus aureus in epidemiology, with a specific focus on virulence mechanisms utilized to evade neutrophils in bacterial pathogenesis. It is important to understand the complex interactions between pathogenic bacteria and neutrophils so that we can disrupt the ability of pathogens to cause disease.
Collapse
Affiliation(s)
- Megan L Lewis
- Department of Physiology & Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Bas G J Surewaard
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada. .,Department of Medical Microbiology, University Medical Centre, Utrecht, Netherlands.
| |
Collapse
|
42
|
Wan TW, Higuchi W, Khokhlova OE, Hung WC, Iwao Y, Wakayama M, Inomata N, Takano T, Lin YT, Peryanova OV, Kojima KK, Salmina AB, Teng LJ, Yamamoto T. Genomic comparison between Staphylococcus aureus GN strains clinically isolated from a familial infection case: IS1272 transposition through a novel inverted repeat-replacing mechanism. PLoS One 2017; 12:e0187288. [PMID: 29117225 PMCID: PMC5678879 DOI: 10.1371/journal.pone.0187288] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 10/17/2017] [Indexed: 01/22/2023] Open
Abstract
A bacterial insertion sequence (IS) is a mobile DNA sequence carrying only the transposase gene (tnp) that acts as a mutator to disrupt genes, alter gene expressions, and cause genomic rearrangements. "Canonical" ISs have historically been characterized by their terminal inverted repeats (IRs), which may form a stem-loop structure, and duplications of a short (non-IR) target sequence at both ends, called target site duplications (TSDs). The IS distributions and virulence potentials of Staphylococcus aureus genomes in familial infection cases are unclear. Here, we determined the complete circular genome sequences of familial strains from a Panton-Valentine leukocidin (PVL)-positive ST50/agr4 S. aureus (GN) infection of a 4-year old boy with skin abscesses. The genomes of the patient strain (GN1) and parent strain (GN3) were rich for "canonical" IS1272 with terminal IRs, both having 13 commonly-existing copies (ce-IS1272). Moreover, GN1 had a newly-inserted IS1272 (ni-IS1272) on the PVL-converting prophage, while GN3 had two copies of ni-IS1272 within the DNA helicase gene and near rot. The GN3 genome also had a small deletion. The targets of ni-IS1272 transposition were IR structures, in contrast with previous "canonical" ISs. There were no TSDs. Based on a database search, the targets for ce-IS1272 were IRs or "non-IRs". IS1272 included a larger structure with tandem duplications of the left (IRL) side sequence; tnp included minor cases of a long fusion form and truncated form. One ce-IS1272 was associated with the segments responsible for immune evasion and drug resistance. Regarding virulence, GN1 expressed cytolytic peptides (phenol-soluble modulin α and δ-hemolysin) and PVL more strongly than some other familial strains. These results suggest that IS1272 transposes through an IR-replacing mechanism, with an irreversible process unlike that of "canonical" transpositions, resulting in genomic variations, and that, among the familial strains, the patient strain has strong virulence potential based on community-associated virulence factors.
Collapse
Affiliation(s)
- Tsai-Wen Wan
- Department of Epidemiology, Genomics, and Evolution, International Medical Education and Research Center, Niigata, Japan
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wataru Higuchi
- Division of Bacteriology, Department of Infectious Disease Control and International Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Olga E. Khokhlova
- Department of Epidemiology, Genomics, and Evolution, International Medical Education and Research Center, Niigata, Japan
- Division of Bacteriology, Department of Infectious Disease Control and International Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Russia-Japan Center of Microbiology, Metagenomics and Infectious Diseases, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
| | - Wei-Chun Hung
- Division of Bacteriology, Department of Infectious Disease Control and International Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Department of Microbiology and Immunology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yasuhisa Iwao
- Department of Epidemiology, Genomics, and Evolution, International Medical Education and Research Center, Niigata, Japan
- Division of Bacteriology, Department of Infectious Disease Control and International Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | | | | | - Tomomi Takano
- Division of Bacteriology, Department of Infectious Disease Control and International Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yu-Tzu Lin
- Department of Epidemiology, Genomics, and Evolution, International Medical Education and Research Center, Niigata, Japan
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Olga V. Peryanova
- Russia-Japan Center of Microbiology, Metagenomics and Infectious Diseases, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
| | - Kenji K. Kojima
- Department of Life Science, National Cheng Kung University, Tainan, Taiwan
- Genetic Information Research Institute (GIRI), Mountain View, CA, United States of America
| | - Alla B. Salmina
- Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical University named after Professor V.F. Vojno-Yasenetsky, Krasnoyarsk, Russia
| | - Lee-Jene Teng
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Tatsuo Yamamoto
- Department of Epidemiology, Genomics, and Evolution, International Medical Education and Research Center, Niigata, Japan
- Division of Bacteriology, Department of Infectious Disease Control and International Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Russia-Japan Center of Microbiology, Metagenomics and Infectious Diseases, Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky, Krasnoyarsk, Russia
- * E-mail:
| |
Collapse
|
43
|
Uribe-Querol E, Rosales C. Control of Phagocytosis by Microbial Pathogens. Front Immunol 2017; 8:1368. [PMID: 29114249 PMCID: PMC5660709 DOI: 10.3389/fimmu.2017.01368] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 10/05/2017] [Indexed: 12/17/2022] Open
Abstract
Phagocytosis is a fundamental process of cells to capture and ingest foreign particles. Small unicellular organisms such as free-living amoeba use this process to acquire food. In pluricellular organisms, phagocytosis is a universal phenomenon that all cells are able to perform (including epithelial, endothelial, fibroblasts, etc.), but some specialized cells (such as neutrophils and macrophages) perform this very efficiently and were therefore named professional phagocytes by Rabinovitch. Cells use phagocytosis to capture and clear all particles larger than 0.5 µm, including pathogenic microorganisms and cellular debris. Phagocytosis involves a series of steps from recognition of the target particle, ingestion of it in a phagosome (phagocytic vacuole), maturation of this phagosome into a phagolysosome, to the final destruction of the ingested particle in the robust antimicrobial environment of the phagolysosome. For the most part, phagocytosis is an efficient process that eliminates invading pathogens and helps maintaining homeostasis. However, several pathogens have also evolved different strategies to prevent phagocytosis from proceeding in a normal way. These pathogens have a clear advantage to perpetuate the infection and continue their replication. Here, we present an overview of the phagocytic process with emphasis on the antimicrobial elements professional phagocytes use. We also summarize the current knowledge on the microbial strategies different pathogens use to prevent phagocytosis either at the level of ingestion, phagosome formation, and maturation, and even complete escape from phagosomes.
Collapse
Affiliation(s)
- Eileen Uribe-Querol
- División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Carlos Rosales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
44
|
Abstract
The life span of a neutrophil is a tightly regulated process as extended survival is beneficial for pathogen elimination and cell death necessary to prevent cytotoxic content release from activated neutrophils at the inflammatory site. Therefore, the control between survival and death must be a dynamic process. We have previously described that proliferating cell nuclear antigen (PCNA) which is known as a nuclear protein pivotal in DNA synthesis, is a key element in controlling neutrophil survival through its association with procaspases. Contrary to the dogma which asserted that PCNA has a strictly nuclear function, in mature neutrophils, PCNA is present exclusively within the cytosol due to its nuclear export at the end of the granulocytic differentiation. More recent studies are consistent with the notion that the cytosolic scaffold of PCNA is aimed at modulating neutrophil fate rather than simply preventing death. Ultimately, targeting neutrophil survival might have important applications not just in the field of immunology and inflammation, but also in hematology and transfusion. The neutrophil emerges as a unique and powerful cellular model to unravel the basic mechanisms governing the cell cycle-independent functions of PCNA and should be considered as a leader of the pack.
Collapse
Affiliation(s)
- Véronique Witko-Sarsat
- INSERM U1016, Paris, France.,Institut Cochin, Faculté de Médecine Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,CNRS UMR 8104, Paris, France.,Center of Excellence, Labex Inflamex, Paris, France
| | - Delphine Ohayon
- INSERM U1016, Paris, France.,Institut Cochin, Faculté de Médecine Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,CNRS UMR 8104, Paris, France.,Center of Excellence, Labex Inflamex, Paris, France
| |
Collapse
|
45
|
Chen X, Li SJ, Ojcius DM, Sun AH, Hu WL, Lin X, Yan J. Mononuclear-macrophages but not neutrophils act as major infiltrating anti-leptospiral phagocytes during leptospirosis. PLoS One 2017; 12:e0181014. [PMID: 28700741 PMCID: PMC5507415 DOI: 10.1371/journal.pone.0181014] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/23/2017] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE To identify the major infiltrating phagocytes during leptospirosis and examine the killing mechanism used by the host to eliminate Leptospira interrogans. METHODS Major infiltrating phagocytes in Leptospira-infected C3H/HeJ mice were detected by immunohistochemistry. Chemokines and vascular endothelial cell adhesion molecules (VECAMs) of Leptospira-infected mice and leptospirosis patients were detected by microarray and immunohistochemistry. Leptospira-phagocytosing and -killing abilities of human or mouse macrophages and neutrophils, and the roles of intracellular ROS, NO and [Ca2+]i in Leptospira-killing process were evaluated by confocal microscopy and spectrofluorimetry. RESULTS Peripheral blood mononuclear-macrophages rather than neutrophils were the main infiltrating phagocytes in the lungs, liver and kidneys of infected mice. Levels of macrophage- but not neutrophil-specific chemokines and VECAMs were significantly increased in the samples from infected mice and patients. All macrophages tested had a higher ability than neutrophils to phagocytose and kill leptospires. Higher ROS and NO levels and [Ca2+]i in the macrophages were involved in killing leptospires. Human macrophages displayed more phagolysosome formation and a stronger leptospire-killing ability to than mouse macrophages. CONCLUSIONS Mononuclear-macrophages but not neutrophils represent the main infiltrating and anti-leptospiral phagocytes during leptospirosis. A lower level of phagosome-lysosome fusion may be responsible for the lower Leptospira-killing ability of human macrophages.
Collapse
Affiliation(s)
- Xu Chen
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Shi-Jun Li
- Guizhou Provincial Center for Disease Control and Prevention, Guiyang, Guizhou, P.R. China
| | - David M. Ojcius
- Department of Biomedical Sciences, University of the Pacific, Arthur Dugoni School of Dentistry, San Francisco, California, United States of America
| | - Ai-Hua Sun
- Faculty of Basic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, P.R. China
| | - Wei-Lin Hu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Xu’ai Lin
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Jie Yan
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
46
|
Mechanism of antagonistic effects of Andrographis paniculata methanolic extract against Staphylococcus aureus. ASIAN PAC J TROP MED 2017; 10:685-695. [DOI: 10.1016/j.apjtm.2017.07.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 05/17/2017] [Accepted: 06/23/2017] [Indexed: 11/19/2022] Open
|
47
|
Galectin-3 Is a Target for Proteases Involved in the Virulence of Staphylococcus aureus. Infect Immun 2017; 85:IAI.00177-17. [PMID: 28438975 DOI: 10.1128/iai.00177-17] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 04/13/2017] [Indexed: 01/21/2023] Open
Abstract
Staphylococcus aureus is a major cause of skin and soft tissue infection. The bacterium expresses four major proteases that are emerging as virulence factors: aureolysin (Aur), V8 protease (SspA), staphopain A (ScpA), and staphopain B (SspB). We hypothesized that human galectin-3, a β-galactoside-binding lectin involved in immune regulation and antimicrobial defense, is a target for these proteases and that proteolysis of galectin-3 is a novel immune evasion mechanism. Indeed, supernatants from laboratory strains and clinical isolates of S. aureus caused galectin-3 degradation. Similar proteolytic capacities were found in Staphylococcus epidermidis isolates but not in Staphylococcus saprophyticus Galectin-3-induced activation of the neutrophil NADPH oxidase was abrogated by bacterium-derived proteolysis of galectin-3, and SspB was identified as the major protease responsible. The impact of galectin-3 and protease expression on S. aureus virulence was studied in a murine skin infection model. In galectin-3+/+ mice, SspB-expressing S. aureus caused larger lesions and resulted in higher bacterial loads than protease-lacking bacteria. No such difference in bacterial load or lesion size was detected in galectin-3-/- mice, which overall showed smaller lesion sizes than the galectin-3+/+ animals. In conclusion, the staphylococcal protease SspB inactivates galectin-3, abrogating its stimulation of oxygen radical production in human neutrophils and increasing tissue damage during skin infection.
Collapse
|
48
|
Guo H, Hall JW, Yang J, Ji Y. The SaeRS Two-Component System Controls Survival of Staphylococcus aureus in Human Blood through Regulation of Coagulase. Front Cell Infect Microbiol 2017; 7:204. [PMID: 28611950 PMCID: PMC5447086 DOI: 10.3389/fcimb.2017.00204] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 05/08/2017] [Indexed: 12/03/2022] Open
Abstract
The SaeRS two-component system plays important roles in regulation of key virulence factors and pathogenicity. In this study, however, we found that the deletion mutation of saeRS enhanced bacterial survival in human blood, whereas complementation of the mutant with SaeRS returned survival to wild-type levels. Moreover, these phenomena were observed in different MRSA genetic background isolates, including HA-MRSA WCUH29, CA-MRSA 923, and MW2. To elucidate which gene(s) regulated by SaeRS contribute to the effect, we conducted a series of complementation studies with selected known SaeRS target genes in trans. We found coagulase complementation abolished the enhanced survival of the SaeRS mutant in human blood. The coa and saeRS deletion mutants exhibited a similar survival phenotype in blood. Intriguingly, heterologous expression of coagulase decreased survival of S. epidermidis in human blood. Further, the addition of recombinant coagulase to blood significantly decreased the survival of S. aureus. Further, analysis revealed staphylococcal resistance to killing by hydrogen peroxide was partially dependent on the presence or absence of coagulase. Furthermore, complementation with coagulase, but not SaeRS, returned saeRS/coa double mutant survival in blood to wild-type levels. These data indicate SaeRS modulates bacterial survival in blood in coagulase-dependent manner. Our results provide new insights into the role of staphylococcal SaeRS and coagulase on bacterial survival in human blood.
Collapse
Affiliation(s)
- Haiyong Guo
- Department of Biological Science, School of Life Science, Jilin Normal UniversitySiping, China.,Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of MinnesotaSt Paul, MN, United States
| | - Jeffrey W Hall
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of MinnesotaSt Paul, MN, United States
| | - Junshu Yang
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of MinnesotaSt Paul, MN, United States
| | - Yinduo Ji
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of MinnesotaSt Paul, MN, United States
| |
Collapse
|
49
|
Rasigade JP. Catching the evader: Can monoclonal antibodies interfere with Staphylococcus aureus immune escape? Virulence 2017; 9:1-4. [PMID: 28441093 PMCID: PMC5955477 DOI: 10.1080/21505594.2017.1320012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
50
|
Neutrophils and Immunity: From Bactericidal Action to Being Conquered. J Immunol Res 2017; 2017:9671604. [PMID: 28299345 PMCID: PMC5337389 DOI: 10.1155/2017/9671604] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/29/2017] [Indexed: 12/19/2022] Open
Abstract
The neutrophil is the major phagocyte and the final effector cell of the innate immunity, with a primary role in the clearance of extracellular pathogens. Using the broad array of cytokines, extracellular traps, and effector molecules as the humoral arm, neutrophils play a crucial role in the host defense against pathogen infections. On the other hand, the pathogen has the capacity to overcome neutrophil-mediated host defense to establish infection causing human disease. Pathogens, such as S. aureus, have the potential to thwart neutrophil chemotaxis and phagocytosis and thereby succeed in evading killing by neutrophils. Furthermore, S. aureus surviving within neutrophils promotes neutrophil cytolysis, resulting in the release of host-derived molecules that promote local inflammation. Here, we provide a detailed overview of the mechanisms by which neutrophils kill the extracellular pathogens and how pathogens evade neutrophils degradation. This review will provide insights that might be useful for the development of novel therapies against infections caused by antibiotic resistant pathogens.
Collapse
|