1
|
Chen J, Wu Y, Zhu Y, Zhang L, Xu Y, Liu Y. Adaptation for Staphylococcus aureus to hosts via insertion mutation in the accessory gene regulator agrC gene: decreased virulence and enhanced persistence capacity. Microbiol Spectr 2025; 13:e0149724. [PMID: 39611824 PMCID: PMC11705864 DOI: 10.1128/spectrum.01497-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/01/2024] [Indexed: 11/30/2024] Open
Abstract
Staphylococcus aureus is an important human pathogen due to its vast array of virulence factors regulated by multiple regulatory mechanisms, including the accessory gene regulator. In this study, two S. aureus strains were simultaneously isolated from the blood of a febrile patient, belonging to the same clone, designated as 23H with a complete hemolytic phenotype, and 23B, exhibiting an incomplete hemolytic phenotype. The genomic comparison between strains 23B and 23H revealed that 23B had a single adenine base insertion at position 923 in the agrC gene, leading to a functional loss of the encoded AgrC. Experimental findings showed that strain 23B had decreased hemolytic activity, lower cytotoxicity against human alveolar epithelial A549 cells and in the Galleria mellonella model, and a reduced ability to survive intracellularly after infecting macrophages, in comparison to 23H. Conversely, 23B exhibited enhanced biofilm formation, greater adherence to A549 cells, and increased persistence in the face of vancomycin and daptomycin treatment. Transcriptomic analysis revealed that 23B upregulated surface protein-encoding genes while simultaneously reducing the expression levels of virulence factors, highlighting the intricate regulatory adjustments facilitating its persistence and reducing pathogenic potential. ATP assay results indicated that 23B maintained elevated ATP levels during the exponential phase yet exhibited reduced levels in the stationary phase when compared with 23H. Our findings suggested that the mutation in the agrC gene of S. aureus results in diminished virulence but markedly enhances persistence. This mutated strain warrants clinical attention because it may lead to treatment failures and persist in patients. IMPORTANCE In clinical antimicrobial therapy, bacterial strains often develop resistance to antimicrobial agents. Additionally, mutations in their gene regulatory networks can increase their persistence, especially in immunocompromised patients. This study identified an insertion mutation in the accessory gene regulator, agrC gene, carried by a Staphylococcus aureus strain isolated from the blood of a febrile patient, leading to the functional loss of AgrC. Further research revealed that despite the reduced virulence of the mutated strain, it significantly bolstered the capacity to adapt and endure within the host during prolonged infections. This was evidenced by increased adhesion and biofilm formation capabilities, development of antimicrobial tolerance, and decreased ATP levels linked to persistence. Therefore, monitoring these mutations in S. aureus is crucial clinically, as they can complicate treatment strategies.
Collapse
Affiliation(s)
- Jiawei Chen
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yun Wu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ying Zhu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Li Zhang
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yingchun Xu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yali Liu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
2
|
Li P, Schulte J, Wurpts G, Hornef MW, Wolz C, Yazdi AS, Burian M. Transcriptional Profiling of Staphylococcus aureus during the Transition from Asymptomatic Nasal Colonization to Skin Colonization/Infection in Patients with Atopic Dermatitis. Int J Mol Sci 2024; 25:9165. [PMID: 39273114 PMCID: PMC11394835 DOI: 10.3390/ijms25179165] [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: 07/24/2024] [Revised: 08/18/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024] Open
Abstract
Staphylococcus aureus acts both as a colonizing commensal bacterium and invasive pathogen. Nasal colonization is associated with an increased risk of infection caused by the identical strain. In patients with atopic dermatitis (AD), the degree of S. aureus colonization is associated with the severity of the disease. Here, we comparatively analyzed the in vivo transcriptional profile of S. aureus colonizing the nose and non-diseased skin (non-lesional skin) as opposed to the diseased skin (lesional skin-defined here as infection) of 12 patients with AD. The transcriptional profile during the asymptomatic colonization of the nose closely resembled that of the lesional skin samples for many of the genes studied, with an elevated expression of the genes encoding adhesion-related proteins and proteases. In addition, the genes that modify and remodel the cell wall and encode proteins that facilitate immune evasion showed increased transcriptional activity. Notably, in a subgroup of patients, the global virulence regulator Agr (accessory gene regulator) and downstream target genes were inactive during nasal colonization but upregulated in the lesional and non-lesional skin samples. Taken together, our results demonstrate a colonization-like transcriptional profile on diseased skin and suggest a role for the peptide quorum sensing system Agr during the transition from asymptomatic nasal colonization to skin colonization/infection.
Collapse
Affiliation(s)
- Peijuan Li
- Department of Dermatology and Allergology, RWTH University Hospital Aachen, D-5207 Aachen, Germany
| | - Julia Schulte
- Department of Dermatology and Allergology, RWTH University Hospital Aachen, D-5207 Aachen, Germany
| | - Gerda Wurpts
- Department of Dermatology and Allergology, RWTH University Hospital Aachen, D-5207 Aachen, Germany
| | - Mathias W Hornef
- Institute of Medical Microbiology, RWTH University Hospital Aachen, D-52074 Aachen, Germany
| | - Christiane Wolz
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tuebingen, D-72076 Tuebingen, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tuebingen, D-72076 Tuebingen, Germany
| | - Amir S Yazdi
- Department of Dermatology and Allergology, RWTH University Hospital Aachen, D-5207 Aachen, Germany
| | - Marc Burian
- Department of Dermatology and Allergology, RWTH University Hospital Aachen, D-5207 Aachen, Germany
| |
Collapse
|
3
|
Dinić M, Burgess JL, Lukić J, Catanuto P, Radojević D, Marjanović J, Verpile R, Thaller SR, Gonzalez T, Golić N, Strahinić I, Tomic-Canic M, Pastar I. Postbiotic lactobacilli induce cutaneous antimicrobial response and restore the barrier to inhibit the intracellular invasion of Staphylococcus aureus in vitro and ex vivo. FASEB J 2024; 38:e23801. [PMID: 39018106 PMCID: PMC11258854 DOI: 10.1096/fj.202400054rr] [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: 01/08/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/18/2024]
Abstract
Intracellular pathogens including Staphylococcus aureus contribute to the non-healing phenotype of chronic wounds. Lactobacilli, well known as beneficial bacteria, are also reported to modulate the immune system, yet their role in cutaneous immunity remains largely unknown. We explored the therapeutic potential of bacteria-free postbiotics, bioactive lysates of lactobacilli, to reduce intracellular S. aureus colonization and promote healing. Fourteen postbiotics derived from various lactobacilli species were screened, and Latilactobacillus curvatus BGMK2-41 was selected for further analysis based on the most efficient ability to reduce intracellular infection by S. aureus diabetic foot ulcer clinical isolate and S. aureus USA300. Treatment of both infected keratinocytes in vitro and infected human skin ex vivo with BGMK2-41 postbiotic cleared S. aureus. Keratinocytes treated in vitro with BGMK2-41 upregulated expression of antimicrobial response genes, of which DEFB4, ANG, and RNASE7 were also found upregulated in treated ex vivo human skin together with CAMP exclusively upregulated ex vivo. Furthermore, BGMK2-41 postbiotic treatment has a multifaceted impact on the wound healing process. Treatment of keratinocytes stimulated cell migration and the expression of tight junction proteins, while in ex vivo human skin BGMK2-41 increased expression of anti-inflammatory cytokine IL-10, promoted re-epithelialization, and restored the epidermal barrier via upregulation of tight junction proteins. Together, this provides a potential therapeutic approach for persistent intracellular S. aureus infections.
Collapse
Affiliation(s)
- Miroslav Dinić
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Jamie L. Burgess
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Molecular and Cellular Pharmacology Graduate Program, University of Miami Miller School of Medicine, Miami FL, USA
| | - Jovanka Lukić
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Paola Catanuto
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Dušan Radojević
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Jelena Marjanović
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rebecca Verpile
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Seth R. Thaller
- DeWitt Daughtry Family Department of Surgery, Division of Plastic Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Tammy Gonzalez
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Nataša Golić
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Ivana Strahinić
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Marjana Tomic-Canic
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Molecular and Cellular Pharmacology Graduate Program, University of Miami Miller School of Medicine, Miami FL, USA
| | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
4
|
Loffredo M, Casciaro B, Bellavita R, Troiano C, Brancaccio D, Cappiello F, Merlino F, Galdiero S, Fabrizi G, Grieco P, Stella L, Carotenuto A, Mangoni ML. Strategic Single-Residue Substitution in the Antimicrobial Peptide Esc(1-21) Confers Activity against Staphylococcus aureus, Including Drug-Resistant and Biofilm Phenotype. ACS Infect Dis 2024; 10:2403-2418. [PMID: 38848266 PMCID: PMC11250030 DOI: 10.1021/acsinfecdis.4c00130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/09/2024]
Abstract
Staphylococcus aureus, a bacterium resistant to multiple drugs, is a significant cause of illness and death worldwide. Antimicrobial peptides (AMPs) provide an excellent potential strategy to cope with this threat. Recently, we characterized a derivative of the frog-skin AMP esculentin-1a, Esc(1-21) (1) that is endowed with potent activity against Gram-negative bacteria but poor efficacy against Gram-positive strains. In this study, three analogues of peptide 1 were designed by replacing Gly8 with α-aminoisobutyric acid (Aib), Pro, and dPro (2-4, respectively). The single substitution Gly8 → Aib8 in peptide 2 makes it active against the planktonic form of Gram-positive bacterial strains, especially Staphylococcus aureus, including multidrug-resistant clinical isolates, with an improved biostability without resulting in cytotoxicity to mammalian cells. Moreover, peptide 2 showed a higher antibiofilm activity than peptide 1 against both reference and clinical isolates of S. aureus. Peptide 2 was also able to induce rapid bacterial killing, suggesting a membrane-perturbing mechanism of action. Structural analysis of the most active peptide 2 evidenced that the improved biological activity of peptide 2 is the consequence of a combination of higher biostability, higher α helical content, and ability to reduce membrane fluidity and to adopt a distorted helix, bent in correspondence of Aib8. Overall, this study has shown how a strategic single amino acid substitution is sufficient to enlarge the spectrum of activity of the original peptide 1, and improve its biological properties for therapeutic purposes, thus paving the way to optimize AMPs for the development of new broad-spectrum anti-infective agents.
Collapse
Affiliation(s)
- Maria
Rosa Loffredo
- Department
of Biochemical Sciences, Laboratory Affiliated to Istituto Pasteur
Italia-Fondazione Cenci Bolognetti, Sapienza
University of Rome, 00185 Rome, Italy
| | - Bruno Casciaro
- Department
of Biochemical Sciences, Laboratory Affiliated to Istituto Pasteur
Italia-Fondazione Cenci Bolognetti, Sapienza
University of Rome, 00185 Rome, Italy
| | - Rosa Bellavita
- Department
of Pharmacy, University of Naples “Federico
II”, 80131 Naples, Italy
| | - Cassandra Troiano
- Department
of Chemical Science and Technologies, University
of Rome Tor Vergata, 00133 Rome, Italy
| | - Diego Brancaccio
- Department
of Pharmacy, University of Naples “Federico
II”, 80131 Naples, Italy
| | - Floriana Cappiello
- Department
of Biochemical Sciences, Laboratory Affiliated to Istituto Pasteur
Italia-Fondazione Cenci Bolognetti, Sapienza
University of Rome, 00185 Rome, Italy
| | - Francesco Merlino
- Department
of Pharmacy, University of Naples “Federico
II”, 80131 Naples, Italy
| | - Stefania Galdiero
- Department
of Pharmacy, University of Naples “Federico
II”, 80131 Naples, Italy
| | - Giancarlo Fabrizi
- Department
of Chemistry and Technology of Drugs, “Department of Excellence
2018−2022”, Sapienza University
of Rome, 00185 Rome, Italy
| | - Paolo Grieco
- Department
of Pharmacy, University of Naples “Federico
II”, 80131 Naples, Italy
| | - Lorenzo Stella
- Department
of Chemical Science and Technologies, University
of Rome Tor Vergata, 00133 Rome, Italy
| | - Alfonso Carotenuto
- Department
of Pharmacy, University of Naples “Federico
II”, 80131 Naples, Italy
| | - Maria Luisa Mangoni
- Department
of Biochemical Sciences, Laboratory Affiliated to Istituto Pasteur
Italia-Fondazione Cenci Bolognetti, Sapienza
University of Rome, 00185 Rome, Italy
| |
Collapse
|
5
|
Morgenstern AR, Peterson LF, Arnold KA, Brewer MG. Differentiation of keratinocytes or exposure to type 2 cytokines diminishes S. aureus internalization. mSphere 2024; 9:e0068523. [PMID: 38501828 PMCID: PMC11036805 DOI: 10.1128/msphere.00685-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/26/2024] [Indexed: 03/20/2024] Open
Abstract
Staphylococcus aureus is a leading cause of skin and soft tissue infections. Colonization by this bacterium is increased in individuals with chronic cutaneous diseases such as atopic dermatitis, psoriasis, and bullous pemphigoid. The greater abundance of S. aureus on the skin of subjects with atopic dermatitis in particular has been linked to recurrent cutaneous infections. The primary cell type of the epidermal layer of the skin is the keratinocyte, and it is thought that S. aureus internalized in keratinocytes associates with an increased incidence of skin infections. This study addresses whether keratinocyte differentiation and/or inflammation, two important characteristics altered in cutaneous diseases, influence bacterial internalization. To do this, S. aureus internalization was measured in immortalized and primary keratinocytes that were differentiated using high Ca2+-containing media and/or exposed to cytokines characteristic of atopic dermatitis (IL-4 and IL-13) or psoriasis (IL-17A and IL-22) skin. Our results indicate that S. aureus internalization is uniquely decreased upon keratinocyte differentiation, since this was not observed with another skin-resident bacterium, S. epidermidis. Additionally, treatment with IL-4 + IL-13 diminished bacterial internalization. We interpret this decrease as a mechanism of keratinocyte-based bacterial killing since a similar number of bacterial genomes were detected in cytokine-treated cells, but less viable internalized S. aureus was recovered. Finally, of the receptors reported for S. aureus binding/internalizing into keratinocytes, expression of the α5 component of the α5β1 integrin was in greatest accordance with the number of internalized bacteria in the context of keratinocyte differentiation.IMPORTANCEIndividuals with chronic cutaneous diseases demonstrate heightened susceptibility for severe and recurrent infections from Staphylococcus aureus. What drives this altered susceptibility remains poorly understood. Previous publications have detected S. aureus as deep as the dermal layer of skin in subjects with atopic dermatitis, suggesting that the cutaneous environment of this disease enables deeper bacterial infiltration than occurs in healthy individuals. This observation indicates that S. aureus has greater opportunity to interact with multiple skin cell types in individuals with chronic inflammatory skin diseases. Identifying the characteristics of the skin that influence bacterial internalization, a common method to establish reservoirs and evade the immune response, is critical for our understanding of S. aureus pathogenesis. The significance of this research is the novel identification of epidermal characteristics that influence S. aureus internalization. With this knowledge, methods can be developed to identify patient populations at greater risk for cutaneous infections.
Collapse
Affiliation(s)
| | - Liam F. Peterson
- Department of Pathology & Laboratory Medicine, University of Rochester, Rochester, New York, USA
| | - Kimberly A. Arnold
- Department of Dermatology, University of Rochester, Rochester, New York, USA
| | - Matthew G. Brewer
- Department of Dermatology, University of Rochester, Rochester, New York, USA
| |
Collapse
|
6
|
Bayer J, Becker J, Liu X, Gritsch L, Daiber E, Korn N, Oesterhelt F, Fraunholz M, Weber A, Wolz C. Differential survival of Staphylococcal species in macrophages. Mol Microbiol 2024; 121:470-480. [PMID: 37898563 DOI: 10.1111/mmi.15184] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/05/2023] [Accepted: 10/02/2023] [Indexed: 10/30/2023]
Abstract
Staphylococcus aureus is considered an extracellular pathogen, yet the bacterium is able to survive within and escape from host cells. An agr/sae mutant of strain USA300 is unable to escape from macrophages but can replicate and survive within. We questioned whether such "non-toxic" S. aureus resembles the less pathogenic coagulase-negative Staphylococcal (CoNS) species like S. epidermidis, S. carnosus, S. lugdunensis, S. capitis, S. warneri, or S. pettenkoferi. We show that the CoNS are more efficiently killed in macrophage-like THP-1 cells or in human primary macrophages. Mutations in katA, copL, the regulatory system graRS, or sigB did not impact bacterial survival in THP-1 cells. Deletion of the superoxide dismutases impaired S. aureus survival in primary macrophages but not in THP-1 cells. However, expression of the S. aureus-specific sodM in S. epidermidis was not sufficient to protect this species from being killed. Thus, at least in those cells, better bacterial survival of S. aureus could not be linked to higher protection from ROS. However, "non-toxic" S. aureus was found to be insensitive to pH, whereas most CoNS were protected when phagosomal acidification was inhibited. Thus, species differences are at least partially linked to differences in sensitivity to acidification.
Collapse
Affiliation(s)
- Janina Bayer
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
| | - Janna Becker
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
| | - Xiao Liu
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Tübingen, Germany
| | - Lisa Gritsch
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
| | - Ellen Daiber
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
| | - Natalya Korn
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
| | - Filipp Oesterhelt
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
| | - Martin Fraunholz
- Department of Microbiology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Alexander Weber
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Tübingen, Germany
| | - Christiane Wolz
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
| |
Collapse
|
7
|
Ding X, Robbe-Masselot C, Fu X, Léonard R, Marsac B, Dauriat CJG, Lepissier A, Rytter H, Ramond E, Dupuis M, Euphrasie D, Dubail I, Schimmich C, Qin X, Parraga J, Leite-de-Moraes M, Ferroni A, Chassaing B, Sermet-Gaudelus I, Charbit A, Coureuil M, Jamet A. Airway environment drives the selection of quorum sensing mutants and promote Staphylococcus aureus chronic lifestyle. Nat Commun 2023; 14:8135. [PMID: 38065959 PMCID: PMC10709412 DOI: 10.1038/s41467-023-43863-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Staphylococcus aureus is a predominant cause of chronic lung infections. While the airway environment is rich in highly sialylated mucins, the interaction of S. aureus with sialic acid is poorly characterized. Using S. aureus USA300 as well as clinical isolates, we demonstrate that quorum-sensing dysfunction, a hallmark of S. aureus adaptation, correlates with a greater ability to consume free sialic acid, providing a growth advantage in an air-liquid interface model and in vivo. Furthermore, RNA-seq experiment reveals that free sialic acid triggers transcriptional reprogramming promoting S. aureus chronic lifestyle. To support the clinical relevance of our results, we show the co-occurrence of S. aureus, sialidase-producing microbiota and free sialic acid in the airway of patients with cystic fibrosis. Our findings suggest a dual role for sialic acid in S. aureus airway infection, triggering virulence reprogramming and driving S. aureus adaptive strategies through the selection of quorum-sensing dysfunctional strains.
Collapse
Affiliation(s)
- Xiongqi Ding
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France
| | - Catherine Robbe-Masselot
- Université Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000, Lille, France
| | - Xiali Fu
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France
| | - Renaud Léonard
- Université Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000, Lille, France
| | - Benjamin Marsac
- Université Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000, Lille, France
| | - Charlene J G Dauriat
- INSERM U1016, CNRS UMR8104, Université Paris Cité, Team «Mucosal Microbiota in Chronic Inflammatory Diseases», F75014, Paris, France
| | - Agathe Lepissier
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France
| | - Héloïse Rytter
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France
| | - Elodie Ramond
- Genoscope, UMR8030, Laboratory of Systems & Synthetic Biology (LISSB), Xenome team, F91057, Evry, France
| | - Marion Dupuis
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France
| | - Daniel Euphrasie
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France
| | - Iharilalao Dubail
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France
| | - Cécile Schimmich
- Anses, Laboratory of Animal Health in Normandy, Physiopathology and epidemiology of equine diseases (PhEED), RD 675, F14430, Goustranville, France
| | - Xiaoquan Qin
- Université Paris Cité, Institut de physique du globe de Paris, CNRS, F75005, Paris, France
| | - Jessica Parraga
- Department of Clinical Microbiology, Necker-Enfants Malades Hospital, AP-HP Centre Université de Paris Cité, F75015, Paris, France
| | - Maria Leite-de-Moraes
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France
| | - Agnes Ferroni
- Department of Clinical Microbiology, Necker-Enfants Malades Hospital, AP-HP Centre Université de Paris Cité, F75015, Paris, France
| | - Benoit Chassaing
- INSERM U1016, CNRS UMR8104, Université Paris Cité, Team «Mucosal Microbiota in Chronic Inflammatory Diseases», F75014, Paris, France
| | - Isabelle Sermet-Gaudelus
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France
| | - Alain Charbit
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France
| | - Mathieu Coureuil
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France.
| | - Anne Jamet
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F75015, Paris, France.
- Department of Clinical Microbiology, Necker-Enfants Malades Hospital, AP-HP Centre Université de Paris Cité, F75015, Paris, France.
| |
Collapse
|
8
|
Liu X, Wang Y, Xi R, Guo D, Guo W, Cheng L, Du T, Lu H, Wang P, Duan Y, Zhu J, Li F. Identification of IRF1 as a Novel Pyroptosis-Related Prognostic Biomarker of Atopic Dermatitis. Genet Test Mol Biomarkers 2023; 27:370-383. [PMID: 38156909 DOI: 10.1089/gtmb.2023.0264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024] Open
Abstract
Purpose: The aim of this study was to characterize key biomarkers associated with pyroptosis in atopic dermatitis (AD). Materials and methods: To identify the differentially expressed pyroptosis-related genes (DEPRGs), the gene expression profiles GSE16161 and GSE32924 from the Gene Expression Omnibus (GEO) database were utilized. Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were conducted to determine the potential biological functions and involved pathways. Furthermore, protein-protein interaction network analyses were performed to identify hub genes. The types and proportions of infiltrating immune cells were detected by immune filtration analysis using CIBERSORT. A 12-axis competing endogenous RNA (ceRNA) network was constructed utilizing the miRNet database. Immunohistochemistry (IHC) further validated the differential expression of a key gene IRF1 in the skin tissues collected from AD patients. The collection of skin tissue from human subjects in this study were reviewed and approved by the IRB of Yueyang Integrated Chinese and Western Medicine Hospital (KYSKSB2020-125). Results: The study identified a total of 76 DEPRGs, which were enriched in genes associated with the inflammatory response and immune regulation. There was a higher percentage of activated dendritic cells and a lower percentage of resting mast cells in AD samples. PVT1 expression was associated with upregulation of hub genes including CXCL8, IRF1, MKI67, and TP53 in the ceRNA network and was correlated with activated dendritic cells in AD. As a transcription factor, IRF1 could regulate the production of downstream inflammatory factors. The IHC study revealed that IRF1 was overexpressed in the skin tissues of AD patients, which were consistent with the results of the bioinformatic study. Conclusions: IRF1 and its related genes were identified as key pyroptosis-related biomarkers in AD, which is a crucial pathway in the pathogenesis of AD.
Collapse
Affiliation(s)
- Xin Liu
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi Wang
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ruofan Xi
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dongjie Guo
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wanjun Guo
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Linyan Cheng
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ting Du
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hanzhi Lu
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Peiyao Wang
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanjuan Duan
- Department of Dermatology, Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianyong Zhu
- Department of Pharmacy Research, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fulun Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
9
|
Dernovics Á, Seprényi G, Rázga Z, Ayaydin F, Veréb Z, Megyeri K. Phenol-Soluble Modulin α3 Stimulates Autophagy in HaCaT Keratinocytes. Biomedicines 2023; 11:3018. [PMID: 38002017 PMCID: PMC10669503 DOI: 10.3390/biomedicines11113018] [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: 09/02/2023] [Revised: 10/30/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Phenol-soluble modulins (PSMs) are pore-forming toxins (PFTs) produced by staphylococci. PSMs exert diverse cellular effects, including lytic, pro-apoptotic, pro-inflammatory and antimicrobial actions. Since the effects of PSMs on autophagy have not yet been reported, we evaluated the autophagic activity in HaCaT keratinocytes treated with recombinant PSMα3. METHODS The autophagic flux and levels of autophagic marker proteins were determined using Western blot analysis. Subcellular localization of LC3B and Beclin-1 was investigated using an indirect immunofluorescence assay. The ultrastructural features of control and PSMα3-treated cells were evaluated via transmission electron microscopy. Cytoplasmic acidification was measured via acridine orange staining. Phosphorylation levels of protein kinases, implicated in autophagy regulation, were studied using a phospho-kinase array and Western blot analysis. RESULTS PSMα3 facilitated the intracellular redistribution of LC3B, increased the average number of autophagosomes per cell, promoted the development of acidic vesicular organelles, elevated the levels of LC3B-II, stimulated autophagic flux and triggered a significant decrease in the net autophagic turnover rate. PSMα3 induced the accumulation of autophagosomes/autolysosomes, amphisomes and multilamellar bodies at the 0.5, 6 and 24 h time points, respectively. The phospho-Akt1/2/3 (T308 and S473), and phospho-mTOR (S2448) levels were decreased, whereas the phospho-Erk1/2 (T202/Y204 and T185/Y187) level was increased in PSMα3-treated cells. CONCLUSIONS In HaCaT keratinocytes, PSMα3 stimulates autophagy. The increased autophagic activity elicited by sub-lytic PSM concentrations might be an integral part of the cellular defense mechanisms protecting skin homeostasis.
Collapse
Affiliation(s)
- Áron Dernovics
- Department of Medical Microbiology, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 10., H-6720 Szeged, Hungary;
| | - György Seprényi
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Kossuth L. sgt. 40., H-6724 Szeged, Hungary;
| | - Zsolt Rázga
- Department of Pathology, University of Szeged, Állomás u. 2, H-6720 Szeged, Hungary;
| | - Ferhan Ayaydin
- Hungarian Centre of Excellence for Molecular Medicine (HCEMM) Nonprofit Ltd., Római krt. 21., H-6723 Szeged, Hungary;
- Laboratory of Cellular Imaging, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62., H-6726 Szeged, Hungary
| | - Zoltán Veréb
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, University of Szeged, Korányi Fasor 6, H-6720 Szeged, Hungary;
- Biobank, University of Szeged, H-6720 Szeged, Hungary
- Interdisciplinary Research Development and Innovation Center of Excellence, University of Szeged, H-6720 Szeged, Hungary
| | - Klára Megyeri
- Department of Medical Microbiology, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 10., H-6720 Szeged, Hungary;
| |
Collapse
|
10
|
Gehrke AKE, Giai C, Gómez MI. Staphylococcus aureus Adaptation to the Skin in Health and Persistent/Recurrent Infections. Antibiotics (Basel) 2023; 12:1520. [PMID: 37887220 PMCID: PMC10604630 DOI: 10.3390/antibiotics12101520] [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: 09/06/2023] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/28/2023] Open
Abstract
Staphylococcus aureus is a microorganism with an incredible capability to adapt to different niches within the human body. Approximately between 20 and 30% of the population is permanently but asymptomatically colonized with S. aureus in the nose, and another 30% may carry S. aureus intermittently. It has been established that nasal colonization is a risk factor for infection in other body sites, including mild to severe skin and soft tissue infections. The skin has distinct features that make it a hostile niche for many bacteria, therefore acting as a strong barrier against invading microorganisms. Healthy skin is desiccated; it has a low pH at the surface; the upper layer is constantly shed to remove attached bacteria; and several host antimicrobial peptides are produced. However, S. aureus is able to overcome these defenses and colonize this microenvironment. Moreover, this bacterium can very efficiently adapt to the stressors present in the skin under pathological conditions, as it occurs in patients with atopic dermatitis or suffering chronic wounds associated with diabetes. The focus of this manuscript is to revise the current knowledge concerning how S. aureus adapts to such diverse skin conditions causing persistent and recurrent infections.
Collapse
Affiliation(s)
- Ana-Katharina E. Gehrke
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Departamento de Investigaciones Biomédicas y Biotecnológicas, Universidad Maimónides, Buenos Aires C1405BCK, Argentina;
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1425FQB, Argentina
| | - Constanza Giai
- Instituto de Histología y Embriología de Mendoza, Universidad Nacional de Cuyo—(UNCuyo) CONICET, Mendoza M5502JMA, Argentina;
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza M5502JMA, Argentina
- Facultad de Farmacia y Bioquímica, Universidad Juan Agustín Maza, Mendoza C1006ACC, Argentina
| | - Marisa I. Gómez
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Departamento de Investigaciones Biomédicas y Biotecnológicas, Universidad Maimónides, Buenos Aires C1405BCK, Argentina;
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1425FQB, Argentina
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121A6B, Argentina
| |
Collapse
|
11
|
Zheng P, Liu F, Long J, Jin Y, Chen S, Duan G, Yang H. Latest Advances in the Application of Humanized Mouse Model for Staphylococcus aureus. J Infect Dis 2023; 228:800-809. [PMID: 37392466 DOI: 10.1093/infdis/jiad253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/21/2023] [Accepted: 06/29/2023] [Indexed: 07/03/2023] Open
Abstract
Staphylococcus aureus (S. aureus) is an important pathogen for humans and can cause a wide range of diseases, from mild skin infections, severe osteomyelitis to fatal pneumonia, sepsis, and septicemia. The mouse models have greatly facilitated the development of S. aureus studies. However, due to the substantial differences in immune system between mice and humans, the conventional mouse studies are not predictive of success in humans, in which case humanized mice may overcome this limitation to some extent. Humanized mice can be used to study the human-specific virulence factors produced by S. aureus and the mechanisms by which S. aureus interacts with humans. This review outlined the latest advances in humanized mouse models used in S. aureus studies.
Collapse
Affiliation(s)
- Ping Zheng
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Fang Liu
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Jinzhao Long
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yuefei Jin
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Shuaiyin Chen
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Guangcai Duan
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Haiyan Yang
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
12
|
Kasela M, Ossowski M, Dzikoń E, Ignatiuk K, Wlazło Ł, Malm A. The Epidemiology of Animal-Associated Methicillin-Resistant Staphylococcus aureus. Antibiotics (Basel) 2023; 12:1079. [PMID: 37370398 DOI: 10.3390/antibiotics12061079] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/14/2023] [Accepted: 06/18/2023] [Indexed: 06/29/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) remains an important etiological factor of human and animal infectious diseases, causing significant economic losses not only in human healthcare but also in the large-scale farming sector. The constantly changing epidemiology of MRSA observed globally affects animal welfare and raises concerns for public health. High MRSA colonization rates in livestock raise questions about the meaning of reservoirs and possible transmission pathways, while the prevalence of MRSA colonization and infection rates among companion animals vary and might affect human health in multiple ways. We present the main findings concerning the circulation of animal-associated MRSA (AA-MRSA) in the environment and factors influencing the direction, mechanisms, and routes of its transmission. Studies have shown it that S. aureus is a multi-host bacterial pathogen; however, its adaptation mechanisms enabling it to colonize and infect both animal and human hosts are still rarely discussed. Finally, we elaborate on the most successful strategies and programs applied limiting the circulation of AA-MRSA among animals and humans. Although MRSA strains colonizing animals rarely infect humans, they undergo host-adaptive evolution enabling them to spread and persist in human populations.
Collapse
Affiliation(s)
- Martyna Kasela
- Department of Pharmaceutical Microbiology, Medical University of Lublin, Chodzki Street 1, 20-093 Lublin, Poland
| | - Mateusz Ossowski
- Department of Animal Hygiene and Environmental Hazards, University of Life Sciences in Lublin, Akademicka 13, 20-950 Lublin, Poland
| | - Ewelina Dzikoń
- Student's Scientific Circle, Department of Pharmaceutical Microbiology, Medical University of Lublin, Chodzki Street 1, 20-093 Lublin, Poland
| | - Katarzyna Ignatiuk
- Student's Scientific Circle, Department of Pharmaceutical Microbiology, Medical University of Lublin, Chodzki Street 1, 20-093 Lublin, Poland
| | - Łukasz Wlazło
- Department of Animal Hygiene and Environmental Hazards, University of Life Sciences in Lublin, Akademicka 13, 20-950 Lublin, Poland
| | - Anna Malm
- Department of Pharmaceutical Microbiology, Medical University of Lublin, Chodzki Street 1, 20-093 Lublin, Poland
| |
Collapse
|
13
|
Linz MS, Mattappallil A, Finkel D, Parker D. Clinical Impact of Staphylococcus aureus Skin and Soft Tissue Infections. Antibiotics (Basel) 2023; 12:557. [PMID: 36978425 PMCID: PMC10044708 DOI: 10.3390/antibiotics12030557] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/03/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
The pathogenic bacterium Staphylococcus aureus is the most common pathogen isolated in skin-and-soft-tissue infections (SSTIs) in the United States. Most S. aureus SSTIs are caused by the epidemic clone USA300 in the USA. These infections can be serious; in 2019, SSTIs with S. aureus were associated with an all-cause, age-standardized mortality rate of 0.5 globally. Clinical presentations of S. aureus SSTIs vary from superficial infections with local symptoms to monomicrobial necrotizing fasciitis, which can cause systemic manifestations and may lead to serious complications or death. In order to cause skin infections, S. aureus employs a host of virulence factors including cytolytic proteins, superantigenic factors, cell wall-anchored proteins, and molecules used for immune evasion. The immune response to S. aureus SSTIs involves initial responders such as keratinocytes and neutrophils, which are supported by dendritic cells and T-lymphocytes later during infection. Treatment for S. aureus SSTIs is usually oral therapy, with parenteral therapy reserved for severe presentations; it ranges from cephalosporins and penicillin agents such as oxacillin, which is generally used for methicillin-sensitive S. aureus (MSSA), to vancomycin for methicillin-resistant S. aureus (MRSA). Treatment challenges include adverse effects, risk for Clostridioides difficile infection, and potential for antibiotic resistance.
Collapse
Affiliation(s)
- Matthew S. Linz
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Arun Mattappallil
- Department of Pharmaceutical Services, University Hospital, Newark, NJ 07103, USA
| | - Diana Finkel
- Division of Infectious Diseases, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Dane Parker
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
14
|
Tamai M, Yamazaki Y, Ito T, Nakagawa S, Nakamura Y. Pathogenic role of the staphylococcal accessory gene regulator quorum sensing system in atopic dermatitis. Front Cell Infect Microbiol 2023; 13:1178650. [PMID: 37124047 PMCID: PMC10140505 DOI: 10.3389/fcimb.2023.1178650] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 04/03/2023] [Indexed: 05/02/2023] Open
Abstract
The skin is home to various bacteria, archaea, fungi, and viruses, collectively referred to as the skin microbiota. Patients with certain skin diseases reportedly have unique skin "dysbiosis," a condition involving imbalanced microbiota, suggesting that dysbiosis in the skin may be either causal or a consequence of specific skin diseases. Atopic dermatitis (AD) is the most common allergic skin disease that affects 15-20% of children and 2-10% of adults worldwide. Both intrinsic genetic factors, such as susceptibility to type 2 inflammation or skin barrier dysfunction, and extrinsic environmental factors, such as air pollen and skin microbiota, contribute to AD. Staphylococcus aureus, which does not often colonize the skin of healthy individuals, is commonly identified in the lesional skin of patients with AD and is correlated with the disease flare. However, the role of S. aureus in the pathogenesis of AD has not been elucidated. Here, we discuss the pathological behavior of S. aureus, focusing on accessory gene regulator (Agr) quorum sensing, which is a fundamental bacterial cell-to-cell interaction mechanism that affects the behavior of S. aureus and other members of the microbial community. Importantly, beyond bacteria-bacteria interactions, the Agr quorum sensing system also regulates various virulence factors, which induce type 2 and IL-17-dependent skin inflammation in the host. Furthermore, the colonization of Agr-positive S. aureus in early life accelerates the development of pediatric AD. Finally, we aim to highlight the current efforts to establish novel therapeutic methods to ameliorate or prevent AD through Agr-targeted intervention.
Collapse
Affiliation(s)
- Masakazu Tamai
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuriko Yamazaki
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
- Cutaneous Allergy and Host Defense, Immunology Frontier Research Center, Osaka University, Osaka, Japan
- *Correspondence: Yuumi Nakamura, ; Yuriko Yamazaki,
| | - Tomoka Ito
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Seitaro Nakagawa
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Yuumi Nakamura
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
- Cutaneous Allergy and Host Defense, Immunology Frontier Research Center, Osaka University, Osaka, Japan
- *Correspondence: Yuumi Nakamura, ; Yuriko Yamazaki,
| |
Collapse
|
15
|
Phenotypic and molecular detection of biofilm formation in clinical methicillin-resistant Staphylococcus aureus isolates from Malaysia. JOURNAL OF TAIBAH UNIVERSITY FOR SCIENCE 2022. [DOI: 10.1080/16583655.2022.2147387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
16
|
Peng G, Tsukamoto S, Ikutama R, Le Thanh Nguyen H, Umehara Y, Trujillo-Paez JV, Yue H, Takahashi M, Ogawa T, Kishi R, Tominaga M, Takamori K, Kitaura J, Kageyama S, Komatsu M, Okumura K, Ogawa H, Ikeda S, Niyonsaba F. Human-β-defensin-3 attenuates atopic dermatitis-like inflammation through autophagy activation and the aryl hydrocarbon receptor signaling pathway. J Clin Invest 2022; 132:156501. [PMID: 35834333 PMCID: PMC9435650 DOI: 10.1172/jci156501] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 07/12/2022] [Indexed: 01/18/2023] Open
Abstract
Human β-defensin-3 (hBD-3) exhibits antimicrobial and immunomodulatory activities; however, its contribution to autophagy regulation remains unclear, and the role of autophagy in the regulation of the epidermal barrier in atopic dermatitis (AD) is poorly understood. Here, keratinocyte autophagy was restrained in the skin lesions of patients with AD and murine models of AD. Interestingly, hBD-3 alleviated the IL-4– and IL-13–mediated impairment of the tight junction (TJ) barrier through keratinocyte autophagy activation, which involved aryl hydrocarbon receptor (AhR) signaling. While autophagy deficiency impaired the epidermal barrier and exacerbated inflammation, hBD-3 attenuated skin inflammation and enhanced the TJ barrier in AD. Importantly, hBD-3–mediated improvement of the TJ barrier was abolished in autophagy-deficient AD mice and in AhR-suppressed AD mice, suggesting a role for hBD-3–mediated autophagy in the regulation of the epidermal barrier and inflammation in AD. Thus, autophagy contributes to the pathogenesis of AD, and hBD-3 could be used for therapeutic purposes.
Collapse
Affiliation(s)
- Ge Peng
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Saya Tsukamoto
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Risa Ikutama
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hai Le Thanh Nguyen
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshie Umehara
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Juan V Trujillo-Paez
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hainan Yue
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Miho Takahashi
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takasuke Ogawa
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ryoma Kishi
- Institute for Environmental and Gender Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan
| | - Mitsutoshi Tominaga
- Institute for Environmental and Gender Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan
| | - Kenji Takamori
- Institute for Environmental and Gender Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan
| | - Jiro Kitaura
- Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shun Kageyama
- Department of Physiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masaaki Komatsu
- Department of Physiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ko Okumura
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hideoki Ogawa
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shigaku Ikeda
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - François Niyonsaba
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
17
|
Exploring the Role of Staphylococcus aureus in Inflammatory Diseases. Toxins (Basel) 2022; 14:toxins14070464. [PMID: 35878202 PMCID: PMC9318596 DOI: 10.3390/toxins14070464] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/23/2022] [Accepted: 07/01/2022] [Indexed: 02/04/2023] Open
Abstract
Staphylococcus aureus is a very common Gram-positive bacterium, and S. aureus infections play an extremely important role in a variety of diseases. This paper describes the types of virulence factors involved, the inflammatory cells activated, the process of host cell death, and the associated diseases caused by S. aureus. S. aureus can secrete a variety of enterotoxins and other toxins to trigger inflammatory responses and activate inflammatory cells, such as keratinocytes, helper T cells, innate lymphoid cells, macrophages, dendritic cells, mast cells, neutrophils, eosinophils, and basophils. Activated inflammatory cells can express various cytokines and induce an inflammatory response. S. aureus can also induce host cell death through pyroptosis, apoptosis, necroptosis, autophagy, etc. This article discusses S. aureus and MRSA (methicillin-resistant S. aureus) in atopic dermatitis, psoriasis, pulmonary cystic fibrosis, allergic asthma, food poisoning, sarcoidosis, multiple sclerosis, and osteomyelitis. Summarizing the pathogenic mechanism of Staphylococcus aureus provides a basis for the targeted treatment of Staphylococcus aureus infection.
Collapse
|
18
|
Mechanisms and Implications of Bacterial Invasion across the Human Skin Barrier. Microbiol Spectr 2022; 10:e0274421. [PMID: 35532353 PMCID: PMC9241919 DOI: 10.1128/spectrum.02744-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Atopic dermatitis (AD) is associated with a deficiency of skin lipids, increased populations of Staphylococcus aureus in the microbiome, and structural defects in the stratum corneum (SC), the outermost layer of human skin. However, the pathogenesis of AD is ambiguous, as it is unclear whether observed changes are the result of AD or contribute to the pathogenesis of the disease. Previous studies have shown that S. aureus is capable of permeating across isolated human SC tissue when lipids are depleted to levels consistent with AD conditions. In this study, we expand upon this discovery to determine the mechanisms and implications of bacterial penetration into the SC barrier. Specifically, we establish if bacteria are permeating intercellularly or employing a combination of both inter- and intracellular travel. The mechanical implications of bacterial invasion, lipid depletion, and media immersion are also evaluated using a newly developed, physiologically relevant, temperature-controlled drip chamber. Results reveal for the first time that S. aureus can be internalized by corneocytes, indicating transcellular movement through the tissue during permeation, consistent with previous theoretical models. S. aureus also degrades the mechanical integrity of human SC, particularly when the tissue is partially depleted of lipids. These observed mechanical changes are likely the cause of broken or ruptured tissue seen as exudative lesions in AD flares. This work further highlights the necessity of lipids in skin microbial barrier function. IMPORTANCE Millions of people suffer from the chronic inflammatory skin disease atopic dermatitis (AD), whose symptoms are associated with a deficiency of skin lipids that exhibit antimicrobial functions and increased populations of the opportunistic pathogen Staphylococcus aureus. However, the pathogenesis of AD is ambiguous, and it remains unclear if these observed changes are merely the result of AD or contribute to the pathogenesis of the disease. In this article, we demonstrate the necessity of skin lipids in preventing S. aureus from penetrating the outermost barrier of human skin, thereby causing a degradation in tissue integrity. This bacterial permeation into the viable epidermis could act as an inflammatory trigger of the disease. When coupled with delipidated AD tissue conditions, bacterial permeation can also explain increased tissue fragility, potentially causing lesion formation in AD patients that results in further enhancing bacterial permeability across the stratum corneum and the development of chronic conditions.
Collapse
|
19
|
Ramond E, Lepissier A, Ding X, Bouvier C, Tan X, Euphrasie D, Monbernard P, Dupuis M, Saubaméa B, Nemazanyy I, Nassif X, Ferroni A, Sermet-Gaudelus I, Charbit A, Coureuil M, Jamet A. Lung-adapted Staphylococcus aureus isolates with dysfunctional agr system trigger a proinflammatory response. J Infect Dis 2022; 226:1276-1285. [PMID: 35524969 DOI: 10.1093/infdis/jiac191] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/05/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Staphylococcus aureus (Sa) dominates the lung microbiota of Cystic Fibrosis (CF) children and persistent clones are able to establish chronic infection for years, having a direct deleterious impact on lung function. However, in this context, the exact contribution of Sa to the decline in respiratory function in CF children is not elucidated. METHODS To investigate the contribution of persistent S. aureus clones in CF disease, we undertook the analysis of sequential isogenic isolates recovered from 15 young CF patients. RESULTS Using an Air-Liquid infection model, we observed a strong correlation between Sa adaption in the lung (late isolates), low toxicity and pro-inflammatory cytokine secretion. Conversely, early isolates appeared to be highly cytotoxic but did not promote cytokine secretion. We found that cytokine secretion was dependent on Staphylococcal protein A (Spa), which was selectively expressed in late compared to early isolates as a consequence of dysfunctional agr quorum-sensing system. Finally, we demonstrated the involvement of TNF-α receptor 1 signaling in the inflammatory response of airway epithelial cells to these lung-adapted Sa isolates. CONCLUSION Our results suggest an unexpected direct role of bacterial lung adaptation in the progression of chronic lung disease by promoting a pro-inflammatory response through acquired agr dysfunction.
Collapse
Affiliation(s)
- Elodie Ramond
- Université de Paris; Faculté de Médecine, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team: Pathogenesis of Systemic Infections, Paris, France
| | - Agathe Lepissier
- Université de Paris; Faculté de Médecine, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team: Epithelial channellopathies, Cystic Fibrosis and other diseases, Paris, France
| | - Xiongqi Ding
- Université de Paris; Faculté de Médecine, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team: Pathogenesis of Systemic Infections, Paris, France
| | - Clémence Bouvier
- Université de Paris; Faculté de Médecine, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team: Pathogenesis of Systemic Infections, Paris, France
| | - Xin Tan
- Université de Paris; Faculté de Médecine, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team: Pathogenesis of Systemic Infections, Paris, France
| | - Daniel Euphrasie
- Université de Paris; Faculté de Médecine, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team: Pathogenesis of Systemic Infections, Paris, France
| | - Pierre Monbernard
- Université de Paris; Faculté de Médecine, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team: Pathogenesis of Systemic Infections, Paris, France
| | - Marion Dupuis
- Université de Paris; Faculté de Médecine, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team: Pathogenesis of Systemic Infections, Paris, France
| | - Bruno Saubaméa
- Cellular and Molecular Imaging facility, INSERM US25, UMS3612 CNRS, Faculté de Pharmacie de Paris, Université de Paris, Paris, France
| | - Ivan Nemazanyy
- Plateforme Etude du métabolisme, Structure Fédérative de Recherche Necker INSERM US24-CNRS UMS 3633, Paris, France
| | - Xavier Nassif
- Université de Paris; Faculté de Médecine, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team: Pathogenesis of Systemic Infections, Paris, France
| | - Agnès Ferroni
- Department of Clinical Microbiology, Necker-Enfants Malades Hospital, AP-HP Centre Université de Paris, Paris, France
| | - Isabelle Sermet-Gaudelus
- Université de Paris; Faculté de Médecine, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team: Epithelial channellopathies, Cystic Fibrosis and other diseases, Paris, France
| | - Alain Charbit
- Université de Paris; Faculté de Médecine, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team: Pathogenesis of Systemic Infections, Paris, France
| | - Mathieu Coureuil
- Université de Paris; Faculté de Médecine, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team: Pathogenesis of Systemic Infections, Paris, France
| | - Anne Jamet
- Université de Paris; Faculté de Médecine, Paris, France.,INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team: Pathogenesis of Systemic Infections, Paris, France.,Department of Clinical Microbiology, Necker-Enfants Malades Hospital, AP-HP Centre Université de Paris, Paris, France
| |
Collapse
|
20
|
The Influence of Antibiotic Resistance on Innate Immune Responses to Staphylococcus aureus Infection. Antibiotics (Basel) 2022; 11:antibiotics11050542. [PMID: 35625186 PMCID: PMC9138074 DOI: 10.3390/antibiotics11050542] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/13/2022] [Accepted: 04/18/2022] [Indexed: 11/16/2022] Open
Abstract
Staphylococcus aureus (S. aureus) causes a broad range of infections and is associated with significant morbidity and mortality. S. aureus produces a diverse range of cellular and extracellular factors responsible for its invasiveness and ability to resist immune attack. In recent years, increasing resistance to last-line anti-staphylococcal antibiotics daptomycin and vancomycin has been observed. Resistant strains of S. aureus are highly efficient in invading a variety of professional and nonprofessional phagocytes and are able to survive inside host cells. Eliciting immune protection against antibiotic-resistant S. aureus infection is a global challenge, requiring both innate and adaptive immune effector mechanisms. Dendritic cells (DC), which sit at the interface between innate and adaptive immune responses, are central to the induction of immune protection against S. aureus. However, it has been observed that S. aureus has the capacity to develop further antibiotic resistance and acquire increased resistance to immunological recognition by the innate immune system. In this article, we review the strategies utilised by S. aureus to circumvent antibiotic and innate immune responses, especially the interaction between S. aureus and DC, focusing on how this relationship is perturbed with the development of antibiotic resistance.
Collapse
|
21
|
Kember M, Grandy S, Raudonis R, Cheng Z. Non-Canonical Host Intracellular Niche Links to New Antimicrobial Resistance Mechanism. Pathogens 2022; 11:pathogens11020220. [PMID: 35215166 PMCID: PMC8876822 DOI: 10.3390/pathogens11020220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/03/2022] [Accepted: 02/05/2022] [Indexed: 12/04/2022] Open
Abstract
Globally, infectious diseases are one of the leading causes of death among people of all ages. The development of antimicrobials to treat infectious diseases has been one of the most significant advances in medical history. Alarmingly, antimicrobial resistance is a widespread phenomenon that will, without intervention, make currently treatable infections once again deadly. In an era of widespread antimicrobial resistance, there is a constant and pressing need to develop new antibacterial drugs. Unraveling the underlying resistance mechanisms is critical to fight this crisis. In this review, we summarize some emerging evidence of the non-canonical intracellular life cycle of two priority antimicrobial-resistant bacterial pathogens: Pseudomonas aeruginosa and Staphylococcus aureus. The bacterial factors that modulate this unique intracellular niche and its implications in contributing to resistance are discussed. We then briefly discuss some recent research that focused on the promises of boosting host immunity as a combination therapy with antimicrobials to eradicate these two particular pathogens. Finally, we summarize the importance of various strategies, including surveillance and vaccines, in mitigating the impacts of antimicrobial resistance in general.
Collapse
|
22
|
Gasdermin D and Beyond - Gasdermin-mediated Pyroptosis in Bacterial Infections. J Mol Biol 2021; 434:167409. [PMID: 34929200 DOI: 10.1016/j.jmb.2021.167409] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 12/21/2022]
Abstract
The discovery of pyroptosis and its subsequent implications in infection and immunity has uncovered a new angle of host-defence against pathogen assault. At its most simple, gasdermin-mediated pyroptosis in bacterial infection would be expected to remove pathogens from the relative safety of the cytosol or pathogen containing vacuole/phagosome whilst inducing a rapid and effective immune response. Differences in gasdermin-mediated pyroptosis between cell types, stimulation conditions, pathogen and even animal species, however, make things more complex. The excessive inflammation associated with the pathogen-induced gasdermin-mediated pyroptosis contributes to a downward spiral in sepsis. With no currently approved effective treatment options for sepsis understanding how gasdermin-mediated pyroptotic pathways are regulated provides an opportunity to identify novel therapeutic candidates against this complex disease. In this review we cover recent advances in the field of gasdermin-mediated pyroptosis with a focus on bacterial infection and sepsis models in the context of humans and other animal species. Importantly we also consider why there is considerable redundancy set into these ancient immune pathways.
Collapse
|
23
|
Pastar I, Sawaya AP, Marjanovic J, Burgess JL, Strbo N, Rivas KE, Wikramanayake TC, Head CR, Stone RC, Jozic I, Stojadinovic O, Kornfeld EY, Kirsner RS, Lev-Tov H, Tomic-Canic M. Intracellular Staphylococcus aureus triggers pyroptosis and contributes to inhibition of healing due to Perforin-2 suppression. J Clin Invest 2021; 131:133727. [PMID: 34730110 DOI: 10.1172/jci133727] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 10/27/2021] [Indexed: 11/17/2022] Open
Abstract
Impaired wound healing associated with recurrent Staphylococcus aureus infection and unresolved inflammation are hallmarks of non-healing diabetic foot ulcers (DFU). Perforin-2, an innate immunity molecule against intracellular bacteria, limits cutaneous infection and dissemination of S. aureus in mice. Here we report the intracellular accumulation of S. aureus in the epidermis of DFU with no clinical signs of infection due to marked suppression of Perforin-2. S. aureus residing within the epidermis of DFU triggers AIM2-inflammasome activation and pyroptosis. These findings were corroborated in mice lacking Perforin-2. The effects of pyroptosis on DFU clinical outcomes were further elucidated in a 4-week longitudinal clinical study in DFU patients undergoing standard of care. Increased AIM2-inflammasome and ASC-pyroptosome coupled with induction of IL-1β were found in non-healing when compared to healing DFU. Our findings reveal novel mechanism that includes Perforin-2 suppression, intracellular S. aureus accumulation and associated induction of pyroptosis that contribute to healing inhibition and prolonged inflammation in patients with DFU.
Collapse
Affiliation(s)
- Irena Pastar
- Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, United States of America
| | - Andrew P Sawaya
- Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, United States of America
| | - Jelena Marjanovic
- Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, United States of America
| | - Jamie L Burgess
- Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, United States of America
| | - Natasa Strbo
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, United States of America
| | - Katelyn E Rivas
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, United States of America
| | - Tongyu C Wikramanayake
- Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, United States of America
| | - Cheyanne R Head
- Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, United States of America
| | - Rivka C Stone
- Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, United States of America
| | - Ivan Jozic
- Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, United States of America
| | - Olivera Stojadinovic
- Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, United States of America
| | - Eran Y Kornfeld
- Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, United States of America
| | - Robert S Kirsner
- University of Miami Miller School of Medicine, Miami, United States of America
| | - Hadar Lev-Tov
- Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, United States of America
| | - Marjana Tomic-Canic
- Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, United States of America
| |
Collapse
|
24
|
Wang M, Fan Z, Han H. Autophagy in Staphylococcus aureus Infection. Front Cell Infect Microbiol 2021; 11:750222. [PMID: 34692566 PMCID: PMC8529010 DOI: 10.3389/fcimb.2021.750222] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/23/2021] [Indexed: 11/23/2022] Open
Abstract
Staphylococcus aureus is an invasive, facultative intracellular pathogen that can colonize niches in various host organisms, making it difficult for the host immune system to completely eliminate. Host autophagy is an intracellular clearance pathway involved in degrading S. aureus. Whereas the accessory gene regulatory system of S. aureus that controls virulence factors could resist the host immune defenses by evading and even utilizing autophagy. This article reviews the interaction between autophagy and S. aureus, providing insights on how to use these mechanisms to improve S. aureus infection control.
Collapse
Affiliation(s)
- Mengyao Wang
- Beijing Key Laboratory of Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China.,National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ziyao Fan
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Hongbing Han
- Beijing Key Laboratory of Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, China.,National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
25
|
Liu C, Gu L, Ding J, Meng Q, Li N, Dai G, Li Q, Wu X. Autophagy in skin barrier and immune-related skin diseases. J Dermatol 2021; 48:1827-1837. [PMID: 34655245 DOI: 10.1111/1346-8138.16185] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/15/2021] [Accepted: 09/24/2021] [Indexed: 12/15/2022]
Abstract
Autophagy is a process which is highly conserved in eukaryotes to degrade or recycle cytoplasmic components through lysosomes to maintain cellular homeostasis. Recent studies have shown that autophagy also plays critical roles in cell apoptosis, inflammation, pathogen clearance, and so on under stressed conditions and thereby has been linked to a variety of human disorders. The skin is the largest organ of the body and serves as the first line of defense against environmental insult. Skin as a nutrient-poor environment requires recycling of limited resources via the autophagy machinery to maintain homeostasis. Therefore, dysregulation of autophagy has been linked to skin diseases. In this review, we describe the molecular machinery and regulation of autophagy, discuss its role in keratinocytes and skin barrier, skin immune cells, and immune-related skin diseases including autoimmune skin disorders, allergic skin diseases, infectious skin disorders, and antitumor immunity against skin tumor. Finally, we highlight the potential of autophagy as a therapeutic target for immune-related skin diseases, and delivery of autophagy-related molecules (such as inducers, inhibitors, or nucleic acid molecules) by virtue of physical materials (such as nanoparticles) or biological materials (such as peptides) to skin topically may obtain clinical benefits in immune-related skin diseases. Moreover, developing autophagy-related gene product-based biomarkers may be promising to diagnose immune-related skin diseases.
Collapse
Affiliation(s)
- Chi Liu
- Department of Geriatrics Center & National Clinical Research Center for Aging and Medicine, Jing'an District Centre Hospital of Shanghai, Fudan University, Shanghai, China.,Department of Cardiology, School of Medicine, Tongji Hospital, Tongji University, Shanghai, China
| | - Lei Gu
- Department of Internal Medicine, Shanghai Shende Hospital, Shanghai, China
| | - Jie Ding
- Department of Gerontology, Jing'an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
| | - Qianchao Meng
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Nan Li
- Department of Geriatrics Center & National Clinical Research Center for Aging and Medicine, Jing'an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
| | - Guifeng Dai
- Department of Geriatrics Center & National Clinical Research Center for Aging and Medicine, Jing'an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
| | - Qinying Li
- Department of Rehabilitation Medicine, Jing'an District Center Hospital of Shanghai, Fudan University, Shanghai, China
| | - Xueyong Wu
- Department of Geriatrics Center & National Clinical Research Center for Aging and Medicine, Jing'an District Centre Hospital of Shanghai, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Ho MKY, Zhang P, Chen X, Xia J, Leung SSY. Bacteriophage endolysins against gram-positive bacteria, an overview on the clinical development and recent advances on the delivery and formulation strategies. Crit Rev Microbiol 2021; 48:303-326. [PMID: 34478359 DOI: 10.1080/1040841x.2021.1962803] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Facing the increasing threat of multi-drug antimicrobial resistance (AMR), humans strive to search for antibiotic drug candidates and antibacterial alternatives from all possible places, from soils in remote areas to deep in the sea. In this "gold rush for antibacterials," researchers turn to the natural enemy of bacterial cells, bacteriophage (phages), and find them a rich source of weapons for AMR bacteria. Endolysins (lysins), the enzymes phages use to break the bacterial cells from within, have been shown to be highly selective and efficient in killing their target bacteria from outside while maintaining a low occurrence of bacterial resistance. In this review, we start with the structures and mechanisms of action of lysins against Gram-positive (GM+) bacteria. The developmental history of lysins is also outlined. Then, we detail the latest preclinical and clinical research on their safety and efficacy against GM+ bacteria, focusing on the formulation strategies of these enzymes. Finally, the challenges and potential hurdles are discussed. Notwithstanding these limitations, the trends in development indicate that the first, approved lysin drugs will be available soon in the near future. Overall, this review presents a timely summary of the current progress on lysins as antibacterial enzymes for AMR GM+ bacteria, and provides a guidebook for biomaterial researchers who are dedicating themselves to the battle against bacterial infections.
Collapse
Affiliation(s)
- Marco Kai Yuen Ho
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong, China
| | - Pengfei Zhang
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong, China
| | - Xi Chen
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiang Xia
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong, China
| | | |
Collapse
|
27
|
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: 2.3] [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
|
28
|
Mat Rani NNI, Mustafa Hussein Z, Mustapa F, Azhari H, Sekar M, Chen XY, Mohd Amin MCI. Exploring the possible targeting strategies of liposomes against methicillin-resistant Staphylococcus aureus (MRSA). Eur J Pharm Biopharm 2021; 165:84-105. [PMID: 33974973 DOI: 10.1016/j.ejpb.2021.04.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 03/26/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022]
Abstract
Multi antibiotic-resistant bacterial infections are on the rise due to the overuse of antibiotics. Methicillin-resistant Staphylococcus aureus (MRSA) is one of the pathogens listed under the category of serious threats where vancomycin remains the mainstay treatment despite the availability of various antibacterial agents. Recently, decreased susceptibility to vancomycin from clinical isolates of MRSA has been reported and has drawn worldwide attention as it is often difficult to overcome and leads to increased medical costs, mortality, and longer hospital stays. Development of antibiotic delivery systems is often necessary to improve bioavailability and biodistribution, in order to reduce antibiotic resistance and increase the lifespan of antibiotics. Liposome entrapment has been used as a method to allow higher drug dosing apart from reducing toxicity associated with drugs. The surface of the liposomes can also be designed and enhanced with drug-release properties, active targeting, and stealth effects to prevent recognition by the mononuclear phagocyte system, thus enhancing its circulation time. The present review aimed to highlight the possible targeting strategies of liposomes against MRSA bacteremia systemically while investigating the magnitude of this effect on the minimum inhibitory concentration level.
Collapse
Affiliation(s)
- Nur Najihah Izzati Mat Rani
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia; Faculty of Pharmacy and Health Sciences, Universiti Kuala Lumpur Royal College of Medicine Perak, 30450 Ipoh, Perak, Malaysia
| | - Zahraa Mustafa Hussein
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Fahimi Mustapa
- Hospital Batu Gajah Jalan Changkat, 31000 Batu Gajah, Perak, Malaysia
| | - Hanisah Azhari
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Mahendran Sekar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Health Sciences, Universiti Kuala Lumpur Royal College of Medicine Perak, 30450 Ipoh, Perak, Malaysia
| | - Xiang Yi Chen
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Mohd Cairul Iqbal Mohd Amin
- Centre for Drug Delivery Technology, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia.
| |
Collapse
|
29
|
Walsh L, Johnson CN, Hill C, Ross RP. Efficacy of Phage- and Bacteriocin-Based Therapies in Combatting Nosocomial MRSA Infections. Front Mol Biosci 2021; 8:654038. [PMID: 33996906 PMCID: PMC8116899 DOI: 10.3389/fmolb.2021.654038] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/07/2021] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus is a pathogen commonly found in nosocomial environments where infections can easily spread - especially given the reduced immune response of patients and large overlap between personnel in charge of their care. Although antibiotics are available to treat nosocomial infections, the increased occurrence of antibiotic resistance has rendered many treatments ineffective. Such is the case for methicillin resistant S. aureus (MRSA), which has continued to be a threat to public health since its emergence. For this reason, alternative treatment technologies utilizing antimicrobials such as bacteriocins, bacteriophages (phages) and phage endolysins are being developed. These antimicrobials provide an advantage over antibiotics in that many have narrow inhibition spectra, enabling treatments to be selected based on the target (pathogenic) bacterium while allowing for survival of commensal bacteria and thus avoiding collateral damage to the microbiome. Bacterial resistance to these treatments occurs less frequently than with antibiotics, particularly in circumstances where combinatory antimicrobial therapies are used. Phage therapy has been well established in Eastern Europe as an effective treatment against bacterial infections. While there are no Randomized Clinical Trials (RCTs) to our knowledge examining phage treatment of S. aureus infections that have completed all trial phases, numerous clinical trials are underway, and several commercial phage preparations are currently available to treat S. aureus infections. Bacteriocins have primarily been used in the food industry for bio-preservation applications. However, the idea of repurposing bacteriocins for human health is an attractive one considering their efficacy against many bacterial pathogens. There are concerns about the ability of bacteriocins to survive the gastrointestinal tract given their proteinaceous nature, however, this obstacle may be overcome by altering the administration route of the therapy through encapsulation, or by bioengineering protease-resistant variants. Obstacles such as enzymatic digestion are less of an issue for topical/local administration, for example, application to the surface of the skin. Bacteriocins have also shown impressive synergistic effects when used in conjunction with other antimicrobials, including antibiotics, which may allow antibiotic-based therapies to be used more sparingly with less resistance development. This review provides an updated account of known bacteriocins, phages and phage endolysins which have demonstrated an impressive ability to kill S. aureus strains. In particular, examples of antimicrobials with the ability to target MRSA strains and their subsequent use in a clinical setting are outlined.
Collapse
Affiliation(s)
- Lauren Walsh
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Crystal N Johnson
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Cork, Ireland
| | - Colin Hill
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - R Paul Ross
- School of Microbiology, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Cork, Ireland
| |
Collapse
|
30
|
Muthukrishnan G, Wallimann A, Rangel-Moreno J, Bentley KLDM, Hildebrand M, Mys K, Kenney HM, Sumrall ET, Daiss JL, Zeiter S, Richards RG, Schwarz EM, Moriarty TF. Humanized Mice Exhibit Exacerbated Abscess Formation and Osteolysis During the Establishment of Implant-Associated Staphylococcus aureus Osteomyelitis. Front Immunol 2021; 12:651515. [PMID: 33815412 PMCID: PMC8012494 DOI: 10.3389/fimmu.2021.651515] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 03/02/2021] [Indexed: 12/17/2022] Open
Abstract
Staphylococcus aureus is the predominant pathogen causing osteomyelitis. Unfortunately, no immunotherapy exists to treat these very challenging and costly infections despite decades of research, and numerous vaccine failures in clinical trials. This lack of success can partially be attributed to an overreliance on murine models where the immune correlates of protection often diverge from that of humans. Moreover, S. aureus secretes numerous immunotoxins with unique tropism to human leukocytes, which compromises the targeting of immune cells in murine models. To study the response of human immune cells during chronic S. aureus bone infections, we engrafted non-obese diabetic (NOD)-scid IL2Rγnull (NSG) mice with human hematopoietic stem cells (huNSG) and analyzed protection in an established model of implant-associated osteomyelitis. The results showed that huNSG mice have increases in weight loss, osteolysis, bacterial dissemination to internal organs, and numbers of Staphylococcal abscess communities (SACs), during the establishment of implant-associated MRSA osteomyelitis compared to NSG controls (p < 0.05). Flow cytometry and immunohistochemistry demonstrated greater human T cell numbers in infected versus uninfected huNSG mice (p < 0.05), and that T-bet+ human T cells clustered around the SACs, suggesting S. aureus-mediated activation and proliferation of human T cells in the infected bone. Collectively, these proof-of-concept studies underscore the utility of huNSG mice for studying an aggressive form of S. aureus osteomyelitis, which is more akin to that seen in humans. We have also established an experimental system to investigate the contribution of specific human T cells in controlling S. aureus infection and dissemination.
Collapse
Affiliation(s)
- Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States
| | - Alexandra Wallimann
- AO Research Institute Davos, Davos, Switzerland.,Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Javier Rangel-Moreno
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Karen L de Mesy Bentley
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States.,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | | | - Karen Mys
- AO Research Institute Davos, Davos, Switzerland
| | - H Mark Kenney
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States
| | | | - John L Daiss
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States
| | | | | | - Edward M Schwarz
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, United States.,Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | | |
Collapse
|
31
|
Soe YM, Bedoui S, Stinear TP, Hachani A. Intracellular Staphylococcus aureus and host cell death pathways. Cell Microbiol 2021; 23:e13317. [PMID: 33550697 DOI: 10.1111/cmi.13317] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/22/2021] [Accepted: 01/23/2021] [Indexed: 12/13/2022]
Abstract
Staphylococcus aureus is a major opportunistic human pathogen that is globally prevalent. Although S. aureus and humans may have co-evolved to the point of commensalism, the bacterium is equipped with virulence factors causing devastating infections. The adoption of an intracellular lifestyle by S. aureus is an important facet of its pathogenesis. Occupying a privileged intracellular compartment permits evasion from the bactericidal actions of host immunity and antibiotics. However, this localization exposes S. aureus to cell-intrinsic processes comprising autophagy, metabolic challenges and clearance mechanisms orchestrated by host programmed cell death pathways (PCDs), including apoptosis, pyroptosis and necroptosis. Mounting evidence suggests that S. aureus deploys pathoadaptive mechanisms that modulate the expression of its virulence factors to prevent elimination through PCD pathways. In this review, we critically analyse the current literature on the interplay between S. aureus virulence factors with the key, intertwined nodes of PCD. We discuss how S. aureus adaptation to the human host plays an essential role in the evasion of PCD, and we consider future directions to study S. aureus-PCD interactions.
Collapse
Affiliation(s)
- Ye Mon Soe
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Sammy Bedoui
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Timothy P Stinear
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Abderrahman Hachani
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
32
|
Missiakas D, Winstel V. Selective Host Cell Death by Staphylococcus aureus: A Strategy for Bacterial Persistence. Front Immunol 2021; 11:621733. [PMID: 33552085 PMCID: PMC7859115 DOI: 10.3389/fimmu.2020.621733] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 12/03/2020] [Indexed: 12/12/2022] Open
Abstract
Host cell death programs are fundamental processes that shape cellular homeostasis, embryonic development, and tissue regeneration. Death signaling and downstream host cell responses are not only critical to guide mammalian development, they often act as terminal responses to invading pathogens. Here, we briefly review and contrast how invading pathogens and specifically Staphylococcus aureus manipulate apoptotic, necroptotic, and pyroptotic cell death modes to establish infection. Rather than invading host cells, S. aureus subverts these cells to produce diffusible molecules that cause death of neighboring hematopoietic cells and thus shapes an immune environment conducive to persistence. The exploitation of cell death pathways by S. aureus is yet another virulence strategy that must be juxtaposed to mechanisms of immune evasion, autophagy escape, and tolerance to intracellular killing, and brings us closer to the true portrait of this pathogen for the design of effective therapeutics and intervention strategies.
Collapse
Affiliation(s)
- Dominique Missiakas
- Howard Taylor Ricketts Laboratory, Department of Microbiology, University of Chicago, Lemont, IL, United States
| | - Volker Winstel
- Research Group Pathogenesis of Bacterial Infections, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
33
|
Casciaro B, Loffredo MR, Cappiello F, Fabiano G, Torrini L, Mangoni ML. The Antimicrobial Peptide Temporin G: Anti-Biofilm, Anti-Persister Activities, and Potentiator Effect of Tobramycin Efficacy Against Staphylococcus aureus. Int J Mol Sci 2020; 21:ijms21249410. [PMID: 33321906 PMCID: PMC7764207 DOI: 10.3390/ijms21249410] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/05/2020] [Accepted: 12/08/2020] [Indexed: 01/10/2023] Open
Abstract
Bacterial biofilms are a serious threat for human health, and the Gram-positive bacterium Staphylococcus aureus is one of the microorganisms that can easily switch from a planktonic to a sessile lifestyle, providing protection from a large variety of adverse environmental conditions. Dormant non-dividing cells with low metabolic activity, named persisters, are tolerant to antibiotic treatment and are the principal cause of recalcitrant and resistant infections, including skin infections. Antimicrobial peptides (AMPs) hold promise as new anti-infective agents to treat such infections. Here for the first time, we investigated the activity of the frog-skin AMP temporin G (TG) against preformed S. aureus biofilm including persisters, as well as its efficacy in combination with tobramycin, in inhibiting S. aureus growth. TG was found to provoke ~50 to 100% reduction of biofilm viability in the concentration range from 12.5 to 100 µM vs ATCC and clinical isolates and to be active against persister cells (about 70–80% killing at 50–100 µM). Notably, sub-inhibitory concentrations of TG in combination with tobramycin were able to significantly reduce S. aureus growth, potentiating the antibiotic power. No critical cytotoxicity was detected when TG was tested in vitro up to 100 µM against human keratinocytes, confirming its safety profile for the development of a new potential anti-infective drug, especially for treatment of bacterial skin infections.
Collapse
Affiliation(s)
- Bruno Casciaro
- Center For Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy
- Correspondence: (B.C.); (M.L.M.); Tel.: +39-0649910838 (M.L.M.)
| | - Maria Rosa Loffredo
- Laboratory Affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (M.R.L.); (F.C.); (G.F.); (L.T.)
| | - Floriana Cappiello
- Laboratory Affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (M.R.L.); (F.C.); (G.F.); (L.T.)
| | - Guendalina Fabiano
- Laboratory Affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (M.R.L.); (F.C.); (G.F.); (L.T.)
| | - Luisa Torrini
- Laboratory Affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (M.R.L.); (F.C.); (G.F.); (L.T.)
| | - Maria Luisa Mangoni
- Laboratory Affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (M.R.L.); (F.C.); (G.F.); (L.T.)
- Correspondence: (B.C.); (M.L.M.); Tel.: +39-0649910838 (M.L.M.)
| |
Collapse
|
34
|
Huitema L, Phillips T, Alexeev V, Tomic-Canic M, Pastar I, Igoucheva O. Intracellular escape strategies of Staphylococcus aureus in persistent cutaneous infections. Exp Dermatol 2020; 30:1428-1439. [PMID: 33179358 DOI: 10.1111/exd.14235] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/19/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022]
Abstract
Pathogenic invasion of Staphylococcus aureus is a major concern in patients with chronic skin diseases like atopic dermatitis (AD), epidermolysis bullosa (EB), or chronic diabetic foot and venous leg ulcers, and can result in persistent and life-threatening chronic non-healing wounds. Staphylococcus aureus is generally recognized as extracellular pathogens. However, S. aureus can also invade, hide and persist in skin cells to contribute to wound chronicity. The intracellular life cycle of S. aureus is currently incompletely understood, although published studies indicate that its intracellular escape strategies play an important role in persistent cutaneous infections. This review provides current scientific knowledge about the intracellular life cycle of S. aureus in skin cells, which can be classified into professional and non-professional antigen-presenting cells, and its strategies to escape adaptive defense mechanisms. First, we discuss phenotypic switch of S. aureus, which affects intracellular routing and degradation. This review also evaluates potential intracellular escape mechanism of S. aureus to avoid intracellular degradation and antigen presentation, preventing an immune response. Furthermore, we discuss potential drug targets that can interfere with the intracellular life cycle of S. aureus. Taken together, this review aimed to increase scientific understanding about the intracellular life cycle of S. aureus into skin cells and its strategies to evade the host immune response, information that is crucial to reduce pathogenic invasion and life-threatening persistence of S. aureus in chronic cutaneous infections.
Collapse
Affiliation(s)
- Leonie Huitema
- Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Taylor Phillips
- Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Vitali Alexeev
- Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Marjana Tomic-Canic
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Olga Igoucheva
- Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
35
|
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: 5.2] [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
|
36
|
Schulz A, Jiang L, de Vor L, Ehrström M, Wermeling F, Eidsmo L, Melican K. Neutrophil Recruitment to Noninvasive MRSA at the Stratum Corneum of Human Skin Mediates Transient Colonization. Cell Rep 2020; 29:1074-1081.e5. [PMID: 31665625 DOI: 10.1016/j.celrep.2019.09.055] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/16/2019] [Accepted: 09/18/2019] [Indexed: 11/16/2022] Open
Abstract
Staphylococcus aureus is a leading cause of skin and soft issue infection, but paradoxically, it also transiently, and often harmlessly, colonizes human skin. An obstacle to understanding this contradiction has been a shortage of in vivo models reproducing the unique structure and immunology of human skin. In this work, we developed a humanized model to study how healthy adult human skin responds to colonizing methicillin-resistant S. aureus (MRSA). We demonstrate the importance of the outer stratum corneum as the major site of bacterial colonization and how noninvasive MRSA adhesion to corneocytes induces a local inflammatory response in underlying skin layers. This signaling recruits neutrophils to the skin, where they control bacterial numbers, mediating transiency in colonization. This work highlights the spatiotemporal aspects of human skin colonization and demonstrates a subclinical inflammatory response to noninvasive MRSA that allows human skin to regulate the bacterial population at its outer surface.
Collapse
Affiliation(s)
- Anette Schulz
- Swedish Medical Nanoscience Center, Department of Neuroscience, Karolinska Institute, Stockholm 171 77, Sweden
| | - Long Jiang
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm 171 77, Sweden
| | - Lisanne de Vor
- Swedish Medical Nanoscience Center, Department of Neuroscience, Karolinska Institute, Stockholm 171 77, Sweden
| | - Marcus Ehrström
- Department of Reconstructive Plastic Surgery, Karolinska University Hospital Solna, Stockholm 171 77, Sweden
| | - Fredrik Wermeling
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm 171 77, Sweden
| | - Liv Eidsmo
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm 171 77, Sweden
| | - Keira Melican
- Swedish Medical Nanoscience Center, Department of Neuroscience, Karolinska Institute, Stockholm 171 77, Sweden.
| |
Collapse
|
37
|
De Maesschalck V, Gutiérrez D, Paeshuyse J, Lavigne R, Briers Y. Advanced engineering of third-generation lysins and formulation strategies for clinical applications. Crit Rev Microbiol 2020; 46:548-564. [PMID: 32886565 DOI: 10.1080/1040841x.2020.1809346] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
One of the possible solutions for the current antibiotic resistance crisis may be found in (often bacteriophage-derived) peptidoglycan hydrolases. The first clinical trials of these natural enzymes, coined here as first-generation lysins, are currently ongoing. Moving beyond natural endolysins with protein engineering established the second generation of lysins. In second-generation lysins, the focus lies on improving antibacterial and biochemical properties such as antimicrobial activity and stability, as well as expanding their activities towards Gram-negative pathogens. However, solutions to particular key challenges regarding clinical applications are only beginning to emerge in the third generation of lysins, in which protein and biochemical engineering efforts focus on improving properties relevant under clinical conditions. In addition, increasingly advanced formulation strategies are developed to increase the bioavailability, antibacterial activity, and half-life, and to reduce pro-inflammatory responses. This review focuses on third-generation and advanced formulation strategies that are developed to treat infections, ranging from topical to systemic applications. Together, these efforts may fully unlock the potential of lysin therapy and will propel it as a true antibiotic alternative or supplement.
Collapse
Affiliation(s)
- Vincent De Maesschalck
- Department of Biosystems, KU Leuven, Leuven, Belgium.,Department of Biotechnology, Ghent University, Gent, Belgium
| | - Diana Gutiérrez
- Department of Biotechnology, Ghent University, Gent, Belgium
| | - Jan Paeshuyse
- Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Rob Lavigne
- Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Yves Briers
- Department of Biotechnology, Ghent University, Gent, Belgium
| |
Collapse
|
38
|
Agarwal Y, Beatty C, Ho S, Thurlow L, Das A, Kelly S, Castronova I, Salunke R, Biradar S, Yeshi T, Richardson A, Bility M. Development of humanized mouse and rat models with full-thickness human skin and autologous immune cells. Sci Rep 2020; 10:14598. [PMID: 32884084 PMCID: PMC7471691 DOI: 10.1038/s41598-020-71548-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 08/13/2020] [Indexed: 12/12/2022] Open
Abstract
The human skin is a significant barrier for protection against pathogen transmission. Rodent models used to investigate human-specific pathogens that target the skin are generated by introducing human skin grafts to immunocompromised rodent strains. Infection-induced immunopathogenesis has been separately studied in humanized rodent models developed with human lymphoid tissue and hematopoietic stem cell transplants. Successful co-engraftment of human skin, autologous lymphoid tissues, and autologous immune cells in a rodent model has not yet been achieved, though it could provide a means of studying the human immune response to infection in the human skin. Here, we introduce the human Skin and Immune System (hSIS)-humanized NOD-scid IL2Rγnull (NSG) mouse and Sprague–Dawley-Rag2tm2hera Il2rγtm1hera (SRG) rat models, co-engrafted with human full-thickness fetal skin, autologous fetal lymphoid tissues, and autologous fetal liver-derived hematopoietic stem cells. hSIS-humanized rodents demonstrate the development of human full-thickness skin, along with autologous lymphoid tissues, and autologous immune cells. These models also support human skin infection following intradermal inoculation with community-associated methicillin-resistant Staphylococcus aureus. The co-engraftment of these human skin and immune system components into a single humanized rodent model could provide a platform for studying human skin infections.
Collapse
Affiliation(s)
- Yash Agarwal
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Cole Beatty
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Sara Ho
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Lance Thurlow
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, USA
| | - Antu Das
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Samantha Kelly
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Isabella Castronova
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Rajeev Salunke
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Shivkumar Biradar
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | | | - Anthony Richardson
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, USA
| | - Moses Bility
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA.
| |
Collapse
|
39
|
Häffner N, Bär J, Dengler Haunreiter V, Mairpady Shambat S, Seidl K, Crosby HA, Horswill AR, Zinkernagel AS. Intracellular Environment and agr System Affect Colony Size Heterogeneity of Staphylococcus aureus. Front Microbiol 2020; 11:1415. [PMID: 32695082 PMCID: PMC7339952 DOI: 10.3389/fmicb.2020.01415] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 06/02/2020] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus causes chronic and relapsing infections, which may be difficult to treat. So-called small colony variants (SCVs) have been associated with chronic infections and their occurrence has been shown to increase under antibiotic pressure, low pH and intracellular localization. In clinics, S. aureus isolated from invasive infections often show a dysfunction in the accessory gene regulator (agr), a major virulence regulatory system in S. aureus. To assess whether intracellular environment and agr function influence SCV formation, an infection model was established using lung epithelial cells and skin fibroblasts. This allowed analyzing intracellular survival and localization of a panel of S. aureus wild type strains and their isogenic agr knock out mutants as well as a natural dysfunctional agr strain by confocal laser scanning microscopy (CLSM). Furthermore, bacterial colonies were quantified after 1, 3, and 5 days of intracellular survival by time-lapse analysis to determine kinetics of colony appearance and SCV formation. Here, we show that S. aureus strains with an agr knock out predominantly resided in a neutral environment, whereas wild type strains and an agr complemented strain resided in an acidic environment. S. aureus agr mutants derived from an intracellular environment showed a higher percentage of SCVs as compared to their corresponding wild type strains. Neutralizing acidic phagolysosomes with chloroquine resulted in a significant reduction of SCVs in S. aureus wild type strain 6850, but not in its agr mutant indicating a pH dependent formation of SCVs in the wild type strain. The in-depth understanding of the interplay between intracellular persistence, agr function and pH should help to identify new therapeutic options facilitating the treatment of chronic S. aureus infections in the future.
Collapse
Affiliation(s)
- Nicola Häffner
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Julian Bär
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Vanina Dengler Haunreiter
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Srikanth Mairpady Shambat
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Kati Seidl
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Heidi A Crosby
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States.,Department of Veterans Affairs Eastern Colorado Health Care System, Denver, CO, United States
| | - Annelies S Zinkernagel
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
40
|
Quaglio D, Corradi S, Erazo S, Vergine V, Berardozzi S, Sciubba F, Cappiello F, Crestoni ME, Ascenzioni F, Imperi F, Delle Monache F, Mori M, Loffredo MR, Ghirga F, Casciaro B, Botta B, Mangoni ML. Structural Elucidation and Antimicrobial Characterization of Novel Diterpenoids from Fabiana densa var. ramulosa. ACS Med Chem Lett 2020; 11:760-765. [PMID: 32435382 DOI: 10.1021/acsmedchemlett.9b00605] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 01/29/2020] [Indexed: 12/20/2022] Open
Abstract
Novel diterpenoids were isolated from the extracts of Fabiana densa var. ramulosa and found to display a selective activity against Gram-positive bacterial strains with negligible cytotoxicity toward human keratinocytes. This study highlighted the role played by the acidic group at C18 of the tetracyclic ent-beyerene scaffold for antibacterial effects and how the length and flexibility of the alkyl chain between the two carbonyl groups are crucial factors to increase the antimicrobial activity of the molecules, supporting the development of natural products from F. densa and their derivatives for treatment of microbial infections.
Collapse
Affiliation(s)
- Deborah Quaglio
- Department of Chemistry and Technology of Drugs, “Department of Excellence 2018−2022”, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Silvia Corradi
- Department of Chemistry and Technology of Drugs, “Department of Excellence 2018−2022”, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
- Center For Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena, 291, 00161 Rome, Italy
| | - Silvia Erazo
- Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Valeria Vergine
- Department of Chemistry and Technology of Drugs, “Department of Excellence 2018−2022”, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Simone Berardozzi
- Department of Chemistry and Applied Biosciences, ETH Zürich, Rämistrasse 101, 8092 Zürich, Switzerland
| | - Fabio Sciubba
- Department of Chemistry, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Floriana Cappiello
- Department of Biochemical Sciences, Laboratory Affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, P.le Aldo Moro 5 00185, Rome, Italy
| | - Maria Elisa Crestoni
- Department of Chemistry and Technology of Drugs, “Department of Excellence 2018−2022”, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Fiorentina Ascenzioni
- Department of Biology and Biotechnology Charles Darwin, Laboratory Affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00161 Rome, Italy
| | | | | | - Mattia Mori
- Department of Biotechnology, Chemistry and Pharmacy, “Department of Excellence 2018−2022”, University of Siena, via Aldo Moro 2, 53100 Siena, Italy
| | - Maria Rosa Loffredo
- Department of Biochemical Sciences, Laboratory Affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, P.le Aldo Moro 5 00185, Rome, Italy
| | - Francesca Ghirga
- Center For Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena, 291, 00161 Rome, Italy
| | - Bruno Casciaro
- Center For Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena, 291, 00161 Rome, Italy
| | - Bruno Botta
- Department of Chemistry and Technology of Drugs, “Department of Excellence 2018−2022”, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| | - Maria Luisa Mangoni
- Department of Biochemical Sciences, Laboratory Affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, P.le Aldo Moro 5 00185, Rome, Italy
| |
Collapse
|
41
|
Research Techniques Made Simple: Mouse Bacterial Skin Infection Models for Immunity Research. J Invest Dermatol 2020; 140:1488-1497.e1. [PMID: 32407714 DOI: 10.1016/j.jid.2020.04.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 01/11/2023]
Abstract
Bacterial skin infections are a major societal health burden and are increasingly difficult to treat owing to the emergence of antibiotic-resistant strains such as community-acquired methicillin-resistant Staphylococcus aureus. Understanding the immunologic mechanisms that provide durable protection against skin infections has the potential to guide the development of immunotherapies and vaccines to engage the host immune response to combat these antibiotic-resistant strains. To this end, mouse skin infection models allow researchers to examine host immunity by investigating the timing, inoculum, route of infection and the causative bacterial species in different wild-type mouse backgrounds as well as in knockout, transgenic, and other types of genetically engineered mouse strains. To recapitulate the various types of human skin infections, many different mouse models have been developed. For example, four models frequently used in dermatological research are based on the route of infection, including (i) subcutaneous infection models, (ii) intradermal infection models, (iii) wound infection models, and (iv) epicutaneous infection models. In this article, we will describe these skin infection models in detail along with their advantages and limitations. In addition, we will discuss how humanized mouse models such as the human skin xenograft on immunocompromised mice might be used in bacterial skin infection research.
Collapse
|
42
|
Lipsky ZW, Marques CNH, German GK. Lipid depletion enables permeation of Staphylococcus aureus bacteria through human stratum corneum. Tissue Barriers 2020; 8:1754706. [PMID: 32338129 PMCID: PMC7549745 DOI: 10.1080/21688370.2020.1754706] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/01/2020] [Accepted: 04/06/2020] [Indexed: 02/01/2023] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory disease that affects approximately 2-5% of adults worldwide. The pathogenesis of AD continues to be a well-debated point of conjecture, with numerous hypotheses having been proposed. AD conditions are associated with increased populations of Staphylococcus aureus and reduced skin lipids. In this study, we evaluate the ability of S. aureus to permeate across human stratum corneum (SC) exhibiting both normal and depleted lipid conditions consistent with AD. This permeation would enable bacteria to interact with underlying viable epidermal cells, which could serve as a trigger for inflammation and disease onset. Our results indicate that permeation of S. aureus through SC exhibiting normal lipid conditions is not statistically significant. However, bacteria can readily permeate through lipid depleted tissue over a 9-d period. These findings suggest that S. aureus may potentially act as the mechanistic cause, rather than merely the result of AD. ABBREVIATIONS AD: Atopic dermatitis; SC: Stratum Corneum; AMP: Antimicrobial peptide; DIW: Deionized water; PDMS: Polydimethylsiloxane; GFP: Green fluorescent protein; BHI: Brain heart infusion medium.
Collapse
Affiliation(s)
- Zachary W. Lipsky
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, NY, USA
| | - Cláudia N. H. Marques
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, NY, USA
- Department of Biological Sciences, Binghamton University, Binghamton, NY, USA
| | - Guy K. German
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, NY, USA
| |
Collapse
|
43
|
Su M, Lyles JT, Petit III RA, Peterson J, Hargita M, Tang H, Solis-Lemus C, Quave CL, Read TD. Genomic analysis of variability in Delta-toxin levels between Staphylococcus aureus strains. PeerJ 2020; 8:e8717. [PMID: 32231873 PMCID: PMC7100594 DOI: 10.7717/peerj.8717] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/10/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND The delta-toxin (δ-toxin) of Staphylococcus aureus is the only hemolysin shown to cause mast cell degranulation and is linked to atopic dermatitis, a chronic inflammatory skin disease. We sought to characterize variation in δ-toxin production across S. aureus strains and identify genetic loci potentially associated with differences between strains. METHODS A set of 124 S. aureus strains was genome-sequenced and δ-toxin levels in stationary phase supernatants determined by high performance liquid chromatography (HPLC). SNPs and kmers were associated with differences in toxin production using four genome-wide association study (GWAS) methods. Transposon mutations in candidate genes were tested for their δ-toxin levels. We constructed XGBoost models to predict toxin production based on genetic loci discovered to be potentially associated with the phenotype. RESULTS The S. aureus strain set encompassed 40 sequence types (STs) in 23 clonal complexes (CCs). δ-toxin production ranged from barely detectable levels to >90,000 units, with a median of >8,000 units. CC30 had significantly lower levels of toxin production than average while CC45 and CC121 were higher. MSSA (methicillin sensitive) strains had higher δ-toxin production than MRSA (methicillin resistant) strains. Through multiple GWAS approaches, 45 genes were found to be potentially associated with toxicity. Machine learning models using loci discovered through GWAS as features were able to predict δ-toxin production (as a high/low binary phenotype) with a precision of .875 and specificity of .990 but recall of .333. We discovered that mutants in the carA gene, encoding the small chain of carbamoyl phosphate synthase, completely abolished toxin production and toxicity in Caenorhabditis elegans. CONCLUSIONS The amount of stationary phase production of the toxin is a strain-specific phenotype likely affected by a complex interaction of number of genes with different levels of effect. We discovered new candidate genes that potentially play a role in modulating production. We report for the first time that the product of the carA gene is necessary for δ-toxin production in USA300. This work lays a foundation for future work on understanding toxin regulation in S. aureus and prediction of phenotypes from genomic sequences.
Collapse
Affiliation(s)
- Michelle Su
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States of America
| | - James T. Lyles
- Center for the Study of Human Health, College of Arts and Sciences, Emory University, Atlanta, GA, United States of America
| | - Robert A. Petit III
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States of America
| | - Jessica Peterson
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States of America
| | - Michelle Hargita
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States of America
| | - Huaqiao Tang
- Center for the Study of Human Health, College of Arts and Sciences, Emory University, Atlanta, GA, United States of America
| | - Claudia Solis-Lemus
- Department of Human Genetics, School of Medicine, Emory University, Atlanta, GA, United States of America
| | - Cassandra L. Quave
- Center for the Study of Human Health, College of Arts and Sciences, Emory University, Atlanta, GA, United States of America
- Department of Dermatology, School of Medicine, Emory University, Atlanta, GA, United States of America
| | - Timothy D. Read
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States of America
- Department of Human Genetics, School of Medicine, Emory University, Atlanta, GA, United States of America
| |
Collapse
|
44
|
Ammanathan V, Vats S, Abraham IM, Manjithaya R. Xenophagy in cancer. Semin Cancer Biol 2020; 66:163-170. [PMID: 32126260 DOI: 10.1016/j.semcancer.2020.02.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 02/17/2020] [Indexed: 12/24/2022]
Abstract
Macroautophagy (herein autophagy) is an intracellular pathway in which cytoplasmic components are captured by double-membrane vesicles (autophagosomes) that eventually fuse with lysosomes to degrade the cargo. Basal levels of autophagy in all eukaryotic cells maintain cellular homeostasis and under conditions of stress, organelles and proteins not essential for survival are degraded. Apart from these functions, cargoes like aggregated proteins, damaged organelles and intracellular pathogens, which are otherwise harmful to cells, are also selectively captured by autophagy and are destined for degradation. In terms of infectious diseases, pathogens are cleared by a specific form of autophagy known as xenophagy. This lysosomal mediated degradation of pathogens also increases the antigen presentation of cells thereby inducing a further immune response. The process of xenophagy provides a broad spectrum of defense mechanism to capture bacterial, viral and protozoan pathogens. However, pathogens have developed ingenious mechanisms to modulate xenophagy to enhance their intracellular survival. Meanwhile, certain pathogens also induce deleterious effects such as chronic inflammation and overexpression of oncogenes in the host system. This over time can increase the susceptibility of the host for tumorigenesis. Hence targeting tumor through anti-microbial mechanisms like xenophagy could be a novel strategy for combinatorial anti-cancer therapy. The recent developments in understanding the role of xenophagy in combating cancer causing pathogens will be discussed in this review.
Collapse
Affiliation(s)
- Veena Ammanathan
- Jawaharlal Nehru Centre for Advanced Scientific Research, 560064, Bangalore, India
| | - Somya Vats
- Jawaharlal Nehru Centre for Advanced Scientific Research, 560064, Bangalore, India
| | - Irine Maria Abraham
- Jawaharlal Nehru Centre for Advanced Scientific Research, 560064, Bangalore, India
| | - Ravi Manjithaya
- Jawaharlal Nehru Centre for Advanced Scientific Research, 560064, Bangalore, India
| |
Collapse
|
45
|
Yu K, Song L, Kang HP, Kwon HK, Back J, Lee FY. Recalcitrant methicillin-resistant Staphylococcus aureus infection of bone cells: Intracellular penetration and control strategies. Bone Joint Res 2020; 9:49-59. [PMID: 32435455 PMCID: PMC7229311 DOI: 10.1302/2046-3758.92.bjr-2019-0131.r1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
AIMS To characterize the intracellular penetration of osteoblasts and osteoclasts by methicillin-resistant Staphylococcus aureus (MRSA) and the antibiotic and detergent susceptibility of MRSA in bone. METHODS Time-lapse confocal microscopy was used to analyze the interaction of MRSA strain USA300 with primary murine osteoblasts and osteoclasts. The effects of early and delayed antibiotic treatments on intracellular and extracellular bacterial colony formation and cell death were quantified. We tested the effects of cefazolin, gentamicin, vancomycin, tetracycline, rifampicin, and ampicillin, as well as agents used in surgical preparation and irrigation. RESULTS MRSA infiltrated bone-resident cells within 15 to 30 minutes. Penetration was most effectively prevented with early (i.e. 30 minutes) antibiotic administration. The combined administration of rifampicin with other antibiotics potentiated their protective effects against MRSA-induced cytotoxicity and most significantly reduced extracellular bacterial bioburden. Gentamicin-containing compounds were most effective in reducing intracellular MRSA bioburden. Of the surgical preparation agents evaluated, betadine reduced in vitro MRSA growth to the greatest extent. CONCLUSION The standard of care for open fractures involves debridement and antibiotics within the first six hours of injury but does not account for the window in which bacteria penetrate cells. Antibiotics must be administered as early as possible after injury or prior to incision to prevent intracellular infestation. Rifampicin can potentiate the capacity of antibiotic regimens to reduce MRSA-induced cytotoxicity.Cite this article: Bone Joint Res. 2020;9(2):49-59.
Collapse
Affiliation(s)
- Kristin Yu
- Department of Orthopaedics and Rehabilitation, Yale University, New Haven, Connecticut, USA
| | - Lee Song
- Department of Orthopaedics, Columbia University, New York, New York, USA
| | - Hyunwoo Paco Kang
- Department of Orthopedic Surgery, University of Southern California, Los Angeles, California, USA
| | - Hyuk-Kwon Kwon
- Department of Orthopaedics and Rehabilitation, Yale University, New Haven, Connecticut, USA
| | - Jungho Back
- Department of Orthopaedics and Rehabilitation, Yale University, New Haven, Connecticut, USA
| | - Francis Y Lee
- Department of Orthopaedics and Rehabilitation, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
46
|
ClpC affects the intracellular survival capacity of Staphylococcus aureus in non-professional phagocytic cells. Sci Rep 2019; 9:16267. [PMID: 31700127 PMCID: PMC6838064 DOI: 10.1038/s41598-019-52731-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 10/22/2019] [Indexed: 12/22/2022] Open
Abstract
Invasion and persistence of bacteria within host cells requires that they adapt to life in an intracellular environment. This adaptation induces bacterial stress through events such as phagocytosis and enhanced nutrient-restriction. During stress, bacteria synthesize a family of proteins known as heat shock proteins (HSPs) to facilitate adaptation and survival. Previously, we determined the Staphylococcus aureus HSP ClpC temporally alters bacterial metabolism and persistence. This led us to hypothesize that ClpC might alter intracellular survival. Inactivation of clpC in S. aureus strain DSM20231 significantly enhanced long-term intracellular survival in human epithelial (HaCaT) and endothelial (EA.hy926) cell lines, without markedly affecting adhesion or invasion. This phenotype was similar across a genetically diverse collection of S. aureus isolates, and was influenced by the toxin/antitoxin encoding locus mazEF. Importantly, MazEF alters mRNA synthesis and/or stability of S. aureus virulence determinants, indicating ClpC may act through the mRNA modulatory activity of MazEF. Transcriptional analyses of total RNAs isolated from intracellular DSM20231 and isogenic clpC mutant cells identified alterations in transcription of α-toxin (hla), protein A (spa), and RNAIII, consistent with the hypothesis that ClpC negatively affects the intracellular survival of S. aureus in non-professional phagocytic cells, via modulation of MazEF and Agr.
Collapse
|
47
|
Staphylococcus aureus small colony variants impair host immunity by activating host cell glycolysis and inducing necroptosis. Nat Microbiol 2019; 5:141-153. [PMID: 31686028 PMCID: PMC10184863 DOI: 10.1038/s41564-019-0597-0] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 09/19/2019] [Indexed: 01/29/2023]
Abstract
Staphylococcus aureus small colony variants (SCVs) are frequently associated with chronic infection, yet they lack expression of many virulence determinants associated with the pathogenicity of wild-type strains. We found that both wild-type S. aureus and a ΔhemB SCV prototype potently activate glycolysis in host cells. Glycolysis and the generation of mitochondrial reactive oxygen species were sufficient to induce necroptosis, a caspase-independent mechanism of host cell death that failed to eradicate S. aureus and instead promoted ΔhemB SCV pathogenicity. To support ongoing glycolytic activity, the ΔhemB SCV induced over a 100-fold increase in the expression of fumC, which encodes an enzyme that catalyses the degradatin of fumarate, an inhibitor of glycolysis. Consistent with fumC-dependent depletion of local fumarate, the ΔhemB SCV failed to elicit trained immunity and protection from a secondary infectious challenge in the skin. The reliance of the S. aureus SCV population on glycolysis accounts for much of its role in the pathogenesis of S. aureus skin infection.
Collapse
|
48
|
Acker KP, Wong Fok Lung T, West E, Craft J, Narechania A, Smith H, O'Brien K, Moustafa AM, Lauren C, Planet PJ, Prince A. Strains of Staphylococcus aureus that Colonize and Infect Skin Harbor Mutations in Metabolic Genes. iScience 2019; 19:281-290. [PMID: 31401351 PMCID: PMC6700416 DOI: 10.1016/j.isci.2019.07.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 12/29/2022] Open
Abstract
Staphylococcus aureus is the most common cause of skin and soft tissue infections, yet the bacterial genetic changes associated with adaptation to human skin are not well characterized. S. aureus strains isolated from patients with chronic skin colonization and intermittent infection were used to determine the staphylococcal genotypes or phenotypes associated with adaptation to human skin. We demonstrate that polymorphisms in metabolic genes, particularly those involved in the tricarboxylic acid cycle, the fumarate-succinate axis, and the generation of terminal electron transporters, are unexpectedly common. These skin-adapted strains activated glycolysis and hypoxia-inducible factor-1α, interleukin (IL)-1β, and IL-18 release from keratinocytes and promoted dermatopathology equivalent to a methicillin-resistant Staphylococcus aureus USA300 control in a murine model of infection. However, in contrast to USA300, a skin-adapted isolate failed to generate protection from a secondary infectious challenge. Within the context of human skin, there appears to be selection for S. aureus metabolic adaptive changes that promote glycolysis and maintain pathogenicity.
Collapse
Affiliation(s)
- Karen P Acker
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Tania Wong Fok Lung
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Emily West
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Joshua Craft
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Apurva Narechania
- Sackler Institute for Comparative Genomics, American Museum of Natural History, New York, NY, USA
| | - Hannah Smith
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Kelsey O'Brien
- Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ahmed M Moustafa
- Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Christine Lauren
- Department of Dermatology and Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Paul J Planet
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Sackler Institute for Comparative Genomics, American Museum of Natural History, New York, NY, USA; Department of Pediatrics, Perelman College of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alice Prince
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
49
|
George SE, Hrubesch J, Breuing I, Vetter N, Korn N, Hennemann K, Bleul L, Willmann M, Ebner P, Götz F, Wolz C. Oxidative stress drives the selection of quorum sensing mutants in the Staphylococcus aureus population. Proc Natl Acad Sci U S A 2019; 116:19145-19154. [PMID: 31488708 PMCID: PMC6754547 DOI: 10.1073/pnas.1902752116] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Quorum sensing (QS) is the central mechanism by which social interactions within the bacterial community control bacterial behavior. QS-negative cells benefit by exploiting public goods produced by the QS-proficient population. Mechanisms to keep the balance between producers and nonproducers within the population are expected but have not been elucidated for peptide-based QS systems in gram-positive pathogens. The Agr system of Staphylococcus aureus comprises the secretion and sensing of an autoinducing peptide to activate its own expression via the response regulator AgrA as well as the expression of a regulatory RNAIII and psmα/psmß coding for phenol-soluble modulins (PSMs). Agr mutants can be monitored on blood agar due to their nonhemolytic phenotype. In vitro evolution and competition experiments show that they readily accumulate in a process that is accelerated by ciprofloxacin, while the wild type (WT) is retained in the population at low numbers. However, agr mutants possess a fitness advantage only under aerobic conditions. Under hypoxia, Agr activity is increased but without the expected fitness cost. The Agr-imposed oxygen-dependent fitness cost is not due to a metabolic burden but due to the reactive oxygen species (ROS)-inducing capacity of the PSMs and RNAIII-regulated factors. Thus, selection of mutants is dictated by the QS system itself. Under aerobic conditions, emergence of agr-negative mutants may provide the population with a fitness advantage while hypoxia favors QS maintenance and even affords increased toxin production. The oxygen-driven tuning of the Agr system might be of importance to provide the pathogen with capabilities crucial for disease progression.
Collapse
Affiliation(s)
- Shilpa Elizabeth George
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen 72076, Germany
| | - Jennifer Hrubesch
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen 72076, Germany
| | - Inga Breuing
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen 72076, Germany
| | - Naisa Vetter
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen 72076, Germany
| | - Natalya Korn
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen 72076, Germany
| | - Katja Hennemann
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen 72076, Germany
| | - Lisa Bleul
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen 72076, Germany
| | - Matthias Willmann
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen 72076, Germany
| | - Patrick Ebner
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen 72076, Germany
| | - Friedrich Götz
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen 72076, Germany
| | - Christiane Wolz
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen 72076, Germany
| |
Collapse
|
50
|
Casciaro B, Calcaterra A, Cappiello F, Mori M, Loffredo MR, Ghirga F, Mangoni ML, Botta B, Quaglio D. Nigritanine as a New Potential Antimicrobial Alkaloid for the Treatment of Staphylococcus aureus-Induced Infections. Toxins (Basel) 2019; 11:toxins11090511. [PMID: 31480508 PMCID: PMC6783983 DOI: 10.3390/toxins11090511] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 08/26/2019] [Accepted: 08/30/2019] [Indexed: 12/20/2022] Open
Abstract
Staphylococcus aureus is a major human pathogen causing a wide range of nosocomial infections including pulmonary, urinary, and skin infections. Notably, the emergence of bacterial strains resistant to conventional antibiotics has prompted researchers to find new compounds capable of killing these pathogens. Nature is undoubtedly an invaluable source of bioactive molecules characterized by an ample chemical diversity. They can act as unique platform providing new scaffolds for further chemical modifications in order to obtain compounds with optimized biological activity. A class of natural compounds with a variety of biological activities is represented by alkaloids, important secondary metabolites produced by a large number of organisms including bacteria, fungi, plants, and animals. In this work, starting from the screening of 39 alkaloids retrieved from a unique in-house library, we identified a heterodimer -carboline alkaloid, nigritanine, with a potent anti-Staphylococcus action. Nigritanine, isolated from Strychnos nigritana, was characterized for its antimicrobial activity against a reference and three clinical isolates of S. aureus. Its potential cytotoxicity was also evaluated at short and long term against mammalian red blood cells and human keratinocytes, respectively. Nigritanine showed a remarkable antimicrobial activity (minimum inhibitory concentration of 128 µM) without being toxic in vitro to both tested cells. The analysis of the antibacterial activity related to the nigritanine scaffold furnished new insights in the structure-activity relationships (SARs) of -carboline, confirming that dimerization improves its antibacterial activity. Taking into account these interesting results, nigritanine can be considered as a promising candidate for the development of new antimicrobial molecules for the treatment of S. aureus-induced infections.
Collapse
Affiliation(s)
- Bruno Casciaro
- Center For Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome 00161, Italy
| | - Andrea Calcaterra
- Department of Chemistry and Technology of Drugs, "Department of Excellence 2018-2022", Sapienza University of Rome, P.le Aldo Moro 5, Rome 00185, Italy
| | - Floriana Cappiello
- Laboratory affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, Rome 00185, Italy
| | - Mattia Mori
- Department of Biotechnology, Chemistry and Pharmacy, "Department of Excellence 2018-2022", University of Siena, via Aldo Moro 2, Siena 53100, Italy
| | - Maria Rosa Loffredo
- Laboratory affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, Rome 00185, Italy
| | - Francesca Ghirga
- Center For Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome 00161, Italy.
| | - Maria Luisa Mangoni
- Laboratory affiliated to Pasteur Italia-Fondazione Cenci Bolognetti, Department of Biochemical Sciences, Sapienza University of Rome, P.le Aldo Moro 5, Rome 00185, Italy.
| | - Bruno Botta
- Department of Chemistry and Technology of Drugs, "Department of Excellence 2018-2022", Sapienza University of Rome, P.le Aldo Moro 5, Rome 00185, Italy
| | - Deborah Quaglio
- Department of Chemistry and Technology of Drugs, "Department of Excellence 2018-2022", Sapienza University of Rome, P.le Aldo Moro 5, Rome 00185, Italy
| |
Collapse
|