1
|
Girma A. Staphylococcus aureus: Current perspectives on molecular pathogenesis and virulence. Cell Surf 2025; 13:100137. [PMID: 39758277 PMCID: PMC11699754 DOI: 10.1016/j.tcsw.2024.100137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/04/2024] [Accepted: 12/07/2024] [Indexed: 01/07/2025] Open
Abstract
Staphylococcus aureus has evolved a sophisticated regulatory system to control its virulence. One of the main roles of this interconnected network is to sense and respond to diverse environmental signals by altering the synthesis of virulence components required for survival in the host, including cell surface adhesins, extracellular enzymes and toxins. The accessory gene regulator (agr), a quorum sensing system that detects the local concentration of a cyclic peptide signaling molecule, is one of the well-studied of these S. aureus regulatory mechanisms. By using this system, S. aureus is able to sense its own population density and translate this information into a specific pattern of gene expression. In addition to Agr, this pathogen senses specific stimuli through various two-component systems and synchronizes responses with alternative sigma factors and cytoplasmic regulators of the SarA protein family. These different regulatory mechanisms combine host and environmental information into a network that guarantees the best possible response of pathogens to changing circumstances. In this article, an overview of the most significant and thoroughly studied regulatory systems of S. aureus is provided, along with a summary of their roles in host interactions.
Collapse
Affiliation(s)
- Abayeneh Girma
- Department of Biology, College of Natural and Computational Science, Mekdela Amba University, P.O. Box 32, Tulu Awuliya, Ethiopia
| |
Collapse
|
2
|
Kuriu A, Ishikawa K, Tsuchiya K, Furuta K, Kaito C. Xenopus laevis as an infection model for human pathogenic bacteria. Infect Immun 2025:e0012625. [PMID: 40310291 DOI: 10.1128/iai.00126-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 04/03/2025] [Indexed: 05/02/2025] Open
Abstract
Animal infection models are essential for understanding bacterial pathogenicity and corresponding host immune responses. In this study, we investigated whether juvenile Xenopus laevis could be used as an infection model for human pathogenic bacteria. Xenopus frogs succumbed to intraperitoneal injection containing the human pathogenic bacteria Staphylococcus aureus, Pseudomonas aeruginosa, and Listeria monocytogenes. In contrast, non-pathogenic bacteria Bacillus subtilis and Escherichia coli did not induce mortality in Xenopus frogs. The administration of appropriate antibiotics suppressed mortality caused by S. aureus and P. aeruginosa. Strains lacking the agr locus, cvfA (rny) gene, or hemolysin genes in S. aureus, LIPI-1-deleted mutant of L. monocytogenes, which attenuate virulence within mammals, exhibited reduced virulence in Xenopus frogs compared with their respective wild-type counterparts. Bacterial distribution analysis revealed that S. aureus persisted in the blood, liver, heart, and muscles of Xenopus frogs until death. These results suggested that intraperitoneal injection of human pathogenic bacteria induces sepsis-like symptoms in Xenopus frogs, supporting their use as a valuable animal model for evaluating antimicrobial efficacy and identifying virulence genes in various human pathogenic bacteria.
Collapse
Affiliation(s)
- Ayano Kuriu
- Division of Molecular Biology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kazuya Ishikawa
- Division of Molecular Biology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kohsuke Tsuchiya
- Division of Immunology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Kazuyuki Furuta
- Division of Molecular Biology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Chikara Kaito
- Division of Molecular Biology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
3
|
Song L, Bai X, Rudramurthy SH, Alam T, Otto M, Matsumoto M. Staphylococcus saprophyticus prevents skin damage by inhibiting Staphylococcus aureus quorum sensing. Sci Rep 2025; 15:13082. [PMID: 40240425 PMCID: PMC12003716 DOI: 10.1038/s41598-025-97044-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
Coagulase-negative staphylococci are prominent skin commensals that play a crucial role in maintaining skin homeostasis and eliminating pathogens. Coagulase-negative staphylococci, such as Staphylococcus caprae, S. hominis, S. simulans, and S. warneri, have been implicated in suppressing skin inflammation by inhibiting S. aureus virulence; however, it remains unclear whether other staphylococcal species, including S. saprophyticus, also prevent S. aureus-induced skin injury. The present study showed that coagulase-negative S. saprophyticus suppresses skin damage by interfering with S. aureus accessory gene regulator (Agr) quorum sensing. To identify novel coagulase-negative staphylococci that inhibit S. aureus virulence, S. aureus was cultured in the presence of culture supernatants from various coagulase-negative Staphylococcus strains. S. saprophyticus culture supernatant significantly inhibited the virulence of S. aureus regulated by the Agr type-I and -II quorum sensing systems. S. saprophyticus secretes cognate autoinducing peptide (AIP) consisting of a 3-amino acid tail and a 5-amino acid thiolactone ring structure similar to that of S. aureus Agr type-I. Synthetic S. saprophyticus AIP mainly inhibited S. aureus Agr type-I and type-II signaling, without affecting pathogen growth. S. aureus Agr type-I virulence was partially inhibited by chimeric peptides in which either the tail or the thiolactone ring of S. aureus AIP was replaced with that of S. saprophyticus AIP. Furthermore, synthetic S. saprophyticus AIP significantly suppressed skin damage, which was associated with reduced pathogen loads, in murine epicutaneous and intradermal S. aureus inoculation models. Our findings demonstrate that commensal S. saprophyticus-derived AIP protects against cutaneous injury by interfering with S. aureus Agr quorum sensing.
Collapse
Affiliation(s)
- Liuying Song
- Department of Pathobiology, College of Veterinary Medicine, The University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Xiaobao Bai
- Department of Pathobiology, College of Veterinary Medicine, The University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Sushma Halekote Rudramurthy
- Department of Pathobiology, College of Veterinary Medicine, The University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Tauqeer Alam
- Department of Pathobiology, College of Veterinary Medicine, The University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Human Bacterial Pathogenesis, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Masanori Matsumoto
- Department of Pathobiology, College of Veterinary Medicine, The University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
4
|
Liu D, Lu Y, Li Z, Pang X, Gao X. Quorum Sensing: Not Just a Bridge Between Bacteria. Microbiologyopen 2025; 14:e70016. [PMID: 40159675 PMCID: PMC11955508 DOI: 10.1002/mbo3.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 02/18/2025] [Accepted: 03/07/2025] [Indexed: 04/02/2025] Open
Abstract
The study of quorum sensing (QS) has gained critical importance, offering insights into bacterial and microorganism communication. QS, regulated by autoinducers, synchronizes collective bacterial behaviors across diverse chemical signals and target genes. This review highlights innovative approaches to regulating QS, emphasizing the potential of quorum quenching and QS inhibitors to mitigate bacterial pathogenicity. These strategies have shown promise in aquaculture and plant resistance, disrupting QS pathways to combat infections. QS also provides opportunities for developing biosensors for early disease detection and preventing biofilm formation, which is critical to overcoming antimicrobial resistance. The applications of QS extend to cancer therapy, with targeted drug delivery systems utilizing QS mechanisms. Advancements in QS regulation, such as the use of nanomaterials, hydrogels, and microplastics, provide novel methods to modulate QS systems. This review explores the latest developments in QS, recognizing its significance in controlling bacterial behavior and its broad impacts on human health and disease management. Integrating these insights into therapeutic strategies and diagnostics represents a pivotal opportunity for medical progress.
Collapse
Affiliation(s)
- Derun Liu
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesjinanChina
| | - Yonglin Lu
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesjinanChina
| | - Ziyun Li
- State Key Laboratory of Microbial TechnologyShandong UniversityQingdaoChina
| | - Xin Pang
- State Key Laboratory of Microbial TechnologyShandong UniversityQingdaoChina
| | - Xueyan Gao
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesjinanChina
- State Key Laboratory of Microbial TechnologyShandong UniversityQingdaoChina
| |
Collapse
|
5
|
Silva-Santana G. Staphylococcus aureus: Dynamics of pathogenicity and antimicrobial-resistance in hospital and community environments - Comprehensive overview. Res Microbiol 2025; 176:104267. [PMID: 39805330 DOI: 10.1016/j.resmic.2025.104267] [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: 10/24/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
This study reviews Staphylococcus aureus, a significant pathogen in both hospital and community-acquired infections, addressing its epidemiology, pathogenesis, and antimicrobial resistance. It highlights virulence mechanisms, such as adhesion factors, toxins, enzymes, and biofilms, which contribute to survival and immune evasion. The spread of resistance occurs through the transfer of mobile genetic elements like SCCmec and genetic mutations. The analysis also compares hospital and community strains, including multidrug-resistant lineages like MRSA, VISA, and VRSA. The study concludes that S. aureus presents a major public health challenge, requiring new therapeutic approaches and preventive strategies.
Collapse
Affiliation(s)
- Giorgio Silva-Santana
- Health Science Center, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro (RJ), Brazil.
| |
Collapse
|
6
|
Ersoy SC, Rose WE, Proctor RA. Bicarbonate Within: A Hidden Modulator of Antibiotic Susceptibility. Antibiotics (Basel) 2025; 14:96. [PMID: 39858381 PMCID: PMC11760860 DOI: 10.3390/antibiotics14010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/11/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Since its standardization, clinical antimicrobial susceptibility testing (AST) has relied upon a standard medium, Mueller-Hinton Broth/Agar (MHB/A), to determine antibiotic resistance. However, this microbiologic medium bears little resemblance to the host milieu, calling into question the physiological relevance of resistance phenotypes it reveals. Recent studies investigating antimicrobial susceptibility in mammalian cell culture media, a more host-mimicking environment, demonstrate that exposure to host factors significantly alters susceptibility profiles. One such factor is bicarbonate, an abundant ion in the mammalian bloodstream/tissues. Importantly, bicarbonate sensitizes methicillin-resistant Staphylococcus aureus (MRSA) to early-generation β-lactams used for the treatment of methicillin-susceptible S. aureus (MSSA). This "NaHCO3-responsive" phenotype is widespread among US MRSA USA300/CC8 bloodstream and skin and soft tissue infection isolates. Translationally, β-lactam therapy has proven effective against NaHCO3-responsive MRSA in both ex vivo simulated endocarditis vegetation (SEV) and in vivo rabbit infective endocarditis (IE) models. Mechanistically, bicarbonate appears to influence mecA expression and PBP2a production/localization, as well as key elements for PBP2a functionality, including the PBP2a chaperone PrsA, components of functional membrane microdomains (FMMs), and wall teichoic acid (WTA) synthesis. The NaHCO3-responsive phenotype highlights the critical role of host factors in shaping antibiotic susceptibility, emphasizing the need to incorporate more physiological conditions into AST protocols.
Collapse
Affiliation(s)
- Selvi C. Ersoy
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Warren E. Rose
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA;
| | - Richard A. Proctor
- Departments of Medicine and Medical Microbiology & Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA;
| |
Collapse
|
7
|
Stevens EJ, Li JD, Hector TE, Drew GC, Hoang K, Greenrod STE, Paterson S, King KC. Within-host competition causes pathogen molecular evolution and perpetual microbiota dysbiosis. THE ISME JOURNAL 2025; 19:wraf071. [PMID: 40244062 PMCID: PMC12066030 DOI: 10.1093/ismejo/wraf071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/31/2025] [Accepted: 04/11/2025] [Indexed: 04/18/2025]
Abstract
Pathogens newly invading a host must compete with resident microbiota. This within-host microbial warfare could lead to more severe disease outcomes or constrain the evolution of virulence. By passaging a widespread pathogen (Staphylococcus aureus) and a natural microbiota community across populations of nematode hosts, we show that the pathogen displaced microbiota and reduced species richness, but maintained its virulence across generations. Conversely, pathogen populations and microbiota passaged in isolation caused more host harm relative to their respective no-host controls. For the evolved pathogens, this increase in virulence was partly mediated by enhanced biofilm formation and expression of the global virulence regulator agr. Whole genome sequencing revealed shifts in the mode of selection from directional (on pathogens evolving in isolation) to fluctuating (on pathogens evolving in host microbiota). This approach also revealed that competitive interactions with the microbiota drove early pathogen genomic diversification. Metagenome sequencing of the passaged microbiota shows that evolution in pathogen-infected hosts caused a significant reduction in community stability (dysbiosis), along with restrictions on the co-existence of some species based on nutrient competition. Our study reveals how microbial competition during novel infection could determine the patterns and processes of evolution with major consequences for host health.
Collapse
Affiliation(s)
- Emily J Stevens
- Department of Biology, University of Oxford, Oxford, Oxfordshire, OX1 3SZ, United Kingdom
- School of Life Sciences, Keele University, Keele, Staffordshire, ST5 5BG, United Kingdom
| | - Jingdi D Li
- Department of Biology, University of Oxford, Oxford, Oxfordshire, OX1 3SZ, United Kingdom
| | - Tobias E Hector
- Department of Biology, University of Oxford, Oxford, Oxfordshire, OX1 3SZ, United Kingdom
| | - Georgia C Drew
- Department of Biology, University of Oxford, Oxford, Oxfordshire, OX1 3SZ, United Kingdom
| | - Kim Hoang
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, 30322, United States
| | - Samuel T E Greenrod
- Department of Biology, University of Oxford, Oxford, Oxfordshire, OX1 3SZ, United Kingdom
| | - Steve Paterson
- Institute of Infection, Veterinary, and Ecological Sciences, University of Liverpool, Liverpool, Wirral, CH64 7TE, United Kingdom
| | - Kayla C King
- Department of Biology, University of Oxford, Oxford, Oxfordshire, OX1 3SZ, United Kingdom
- Department of Zoology, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| |
Collapse
|
8
|
Ingmer H, Leisner JJ, Fulaz S. Forssman and the staphylococcal hemolysins. APMIS 2025; 133:e13459. [PMID: 39188243 PMCID: PMC11669744 DOI: 10.1111/apm.13459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/05/2024] [Indexed: 08/28/2024]
Abstract
Forssman was a Swedish pathologist and microbiologist who, in the 1920s and 1930s conducted a long series of experiments that led to unique insights into surface antigens of blood cells, as well as added to the discrimination of toxins produced by staphylococci that lyse red blood cells. This review takes offset in the studies published by Forssman in APMIS addressing the hemolytic properties of staphylococcal toxins displayed against erythrocytes of animal and human origin. In light of current knowledge, we will discuss the insights we now have and how they may pave the way for curing infections with pathogenic staphylococci, including Staphylococcus aureus.
Collapse
Affiliation(s)
- Hanne Ingmer
- Department of Veterinary and Animal ScienceUniversity of CopenhagenCopenhagenDenmark
| | - Jørgen J. Leisner
- Department of Veterinary and Animal ScienceUniversity of CopenhagenCopenhagenDenmark
| | - Stephanie Fulaz
- Department of Veterinary and Animal ScienceUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
9
|
Cochard B, Ciprani A, Cirillo M, Habre C, Vazquez O, Frizon L, Bracci B, Dayer R, Krzysztofiak A, Ceroni D. Prevalence of Complications Due to Transphyseal Hematogenous Osteomyelitis. J Bone Joint Surg Am 2024; 107:00004623-990000000-01289. [PMID: 39729530 PMCID: PMC11781545 DOI: 10.2106/jbjs.24.00101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2024]
Abstract
BACKGROUND Transphyseal hematogenous osteomyelitis (THO) is a common infectious condition, being present in 25% of patients with hematogenous osteomyelitis. A large proportion of pediatric hematogenous osteomyelitis infections can spread through the growth cartilage and therefore may be potentially responsible for growth disorders, leading to limb-length discrepancy or angular deformities. The purpose of the present study was to identify both the prevalence of complications caused by transphyseal osteomyelitis and factors influencing their occurrence. METHODS The records for all patients who had been treated for THO over a 17-year period at the University Hospitals of Geneva and the Gesù Bambino Hospital in Rome were retrospectively analyzed. Clinical, biological, and bacteriological data were analyzed. Magnetic resonance imaging (MRI) scans were reviewed for all patients to assess the cross-sectional area of growth plate involvement. Restart of growth of the affected physeal cartilage was subsequently monitored by means of iterative radiographic examination. RESULTS From a cohort of 594 patients with hematogenous osteomyelitis, 89 patients (15.0%) were found to have THO. The median age was 84 months (range, 1 to 167 months), with a bimodal distribution and peaks at 30 and 150 months; 59.6% (53) of the 89 patients were male. The lower limbs were most often affected, with the distal tibia and fibula accounting for 47.2% of all cases. Methicillin-sensitive Staphylococcus aureus and Kingella kingae were the most frequently identified pathogens, accounting for 61.8% of the cases. Virulence factors, such as Panton-Valentine leukocidin (PVL), toxic shock syndrome toxin (TSST), and accessory gene regulator (Agr), were recorded in 12.4% of MSSA strains. Transphyseal lesions affected a median of 7.6% (range, 0.79% to 58.2%) of the physeal surface. Complications affecting further growth were noted in 13% of patients with THO. Thrombocytopenia, leukocytosis, and the presence of virulence factors significantly influenced the occurrence of complications. CONCLUSIONS THO affects all age categories of the pediatric population. In the present study, growth disturbance occurred in 13% of cases. The presence of deep thrombocytopenia, leukocytosis, and virulence factors, such as Agr, TSST, and PVL, seems to strongly influence the occurrence of such complications. LEVEL OF EVIDENCE Therapeutic Level IV. See Instructions for Authors for a complete description of levels of evidence.
Collapse
Affiliation(s)
- Blaise Cochard
- Pediatric Orthopaedic Unit, Pediatric Surgery Service, Geneva University Hospitals, Geneva, Switzerland
- Division of Orthopaedic and Trauma Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Asia Ciprani
- Bambino Gesù Children’s Hospital, Immunology and Infectious Diseases University, Rome, Italy
| | - Marco Cirillo
- Department of Imaging, Bambino Gesù Children’s Hospital, Rome, Italy
| | - Céline Habre
- Pediatric Radiology Unit, Geneva Children’s Hospital, Geneva University Hospitals, Geneva, Switzerland
| | - Oscar Vazquez
- Pediatric Orthopaedic Unit, Pediatric Surgery Service, Geneva University Hospitals, Geneva, Switzerland
- Division of Orthopaedic and Trauma Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Louise Frizon
- Department of Pediatrics, Children’s Hospital, Geneva University Hospitals, Geneva, Switzerland
| | - Benedetta Bracci
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome, Radiology Unit, Sant’Andrea University Hospital, Rome, Italy
| | - Romain Dayer
- Pediatric Orthopaedic Unit, Pediatric Surgery Service, Geneva University Hospitals, Geneva, Switzerland
| | - Andrezj Krzysztofiak
- Bambino Gesù Children’s Hospital, Immunology and Infectious Diseases University, Rome, Italy
| | - Dimitri Ceroni
- Pediatric Orthopaedic Unit, Pediatric Surgery Service, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
10
|
Ziegert Z, Dietz M, Hill M, McBride M, Painter E, Elias MH, Staley C. Targeting quorum sensing for manipulation of commensal microbiota. BMC Biotechnol 2024; 24:106. [PMID: 39696328 PMCID: PMC11653937 DOI: 10.1186/s12896-024-00937-3] [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: 08/26/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024] Open
Abstract
Bacteria communicate through the accumulation of autoinducer (AI) molecules that regulate gene expression at critical densities in a process called quorum sensing (QS). Extensive work using simple systems and single strains of bacteria have revealed a role for QS in the regulation of virulence factors and biofilm formation; however, less is known about QS dynamics among communities, especially in vivo. In this review, we summarize the diversity of QS signals as well as their ability to influence "non-target" behaviors among species that have receptors but not synthases for those signals. We highlight host-microbe interactions facilitated by QS and describe cross-talk between QS and the mammalian endocrine and immune systems, as well as host surveillance of QS. Further, we describe emerging evidence for the role of QS in non-infectious, chronic, microbially associated diseases including inflammatory bowel diseases and cancers. Finally, we describe potential therapeutic approaches that involve leveraging QS signals as well as quorum quenching approaches to block signaling in vivo to mitigate deleterious consequences to the host. Ultimately, QS offers a previously underexplored target that may be leveraged for precision modification of the microbiota without deleterious bactericidal consequences.
Collapse
Affiliation(s)
- Zachary Ziegert
- Division of Basic & Translational Research, Department of Surgery, University of Minnesota Medical School, 420 Delaware St, SE MMC 195, Minneapolis, MN, 55455, USA
- BioTechnology Institute, University of Minnesota, St. Paul, MN, 55108, USA
| | - Matthew Dietz
- Division of Basic & Translational Research, Department of Surgery, University of Minnesota Medical School, 420 Delaware St, SE MMC 195, Minneapolis, MN, 55455, USA
- BioTechnology Institute, University of Minnesota, St. Paul, MN, 55108, USA
| | - Max Hill
- Division of Basic & Translational Research, Department of Surgery, University of Minnesota Medical School, 420 Delaware St, SE MMC 195, Minneapolis, MN, 55455, USA
- BioTechnology Institute, University of Minnesota, St. Paul, MN, 55108, USA
| | - Marjais McBride
- Division of Basic & Translational Research, Department of Surgery, University of Minnesota Medical School, 420 Delaware St, SE MMC 195, Minneapolis, MN, 55455, USA
- BioTechnology Institute, University of Minnesota, St. Paul, MN, 55108, USA
| | - Elizabeth Painter
- Division of Basic & Translational Research, Department of Surgery, University of Minnesota Medical School, 420 Delaware St, SE MMC 195, Minneapolis, MN, 55455, USA
- BioTechnology Institute, University of Minnesota, St. Paul, MN, 55108, USA
| | - Mikael H Elias
- BioTechnology Institute, University of Minnesota, St. Paul, MN, 55108, USA
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, St. Paul, MN, 55108, USA
| | - Christopher Staley
- Division of Basic & Translational Research, Department of Surgery, University of Minnesota Medical School, 420 Delaware St, SE MMC 195, Minneapolis, MN, 55455, USA.
- BioTechnology Institute, University of Minnesota, St. Paul, MN, 55108, USA.
| |
Collapse
|
11
|
Ito T, Nakamura Y. The skin barrier and microbiome in infantile atopic dermatitis development: can skincare prevent onset? Int Immunol 2024; 36:579-584. [PMID: 38887075 DOI: 10.1093/intimm/dxae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/17/2024] [Indexed: 06/20/2024] Open
Abstract
Atopic dermatitis (AD), a prevalent Th2-dominant skin disease, involves complex genetic and environmental factors, including mutations in the Filaggrin gene and dysbiosis of skin microbiota characterized by an increased abundance of Staphylococcus aureus. Our recent findings emphasize the pivotal role of the skin barrier's integrity and microbial composition in infantile AD and allergic diseases. Early skin dysbiosis predisposes infants to AD, suggesting targeted skincare practices as a preventive strategy. The effects of skincare interventions, particularly the application of moisturizers with the appropriate molar concentration of ceramides, cholesterol, and fatty acids, play a crucial role in restoring the skin barrier. Notably, our study revealed that appropriate skincare can reduce Streptococcus abundance while supporting Cutibacterium acnes presence, thus directly linking skincare practices to microbial modulation in neonatal skin. Despite the mixed outcomes of previous Randomized Controlled Trials on the efficacy of moisturizers in AD prevention, our research points to the potential of skincare intervention as a primary preventive method against AD by minimizing the impact of genetic and environmental factors. Furthermore, our research supports the notion that early aggressive management of eczema may reduce the incidence of food allergies, highlighting the necessity for multifaceted prevention strategies that address both the skin barrier and immune sensitization. By focusing on repairing the skin barrier and adjusting the skin's microbiome from birth, we propose a novel perspective on preventing infantile AD and allergic diseases, opening new avenues for future studies, and practices in allergy prevention.
Collapse
Affiliation(s)
- Tomoka Ito
- Department of Dermatology, The University of Osaka, Graduate School of Medicine, Osaka 565-0871, Japan
| | - Yuumi Nakamura
- Department of Dermatology, The University of Osaka, Graduate School of Medicine, Osaka 565-0871, Japan
- Cutaneous Allergy and Host Defense, Immunology Frontier Research Center, The University of Osaka, Osaka 565-0871, Japan
| |
Collapse
|
12
|
Su M, Hoang KL, Penley M, Davis MH, Gresham JD, Morran LT, Read TD. Host and antibiotic jointly select for greater virulence in Staphylococcus aureus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.31.610628. [PMID: 39257827 PMCID: PMC11383984 DOI: 10.1101/2024.08.31.610628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Widespread antibiotic usage has resulted in the rapid evolution of drug-resistant bacterial pathogens and poses significant threats to public health. Resolving how pathogens respond to antibiotics under different contexts is critical for understanding disease emergence and evolution going forward. The impact of antibiotics has been demonstrated most directly through in vitro pathogen passaging experiments. Independent from antibiotic selection, interactions with hosts have also altered the evolutionary trajectories and fitness landscapes of pathogens, shaping infectious disease outcomes. However, it is unclear how interactions between hosts and antibiotics impact the evolution of pathogen virulence. Here, we evolved and re-sequenced Staphylococcus aureus, a major bacterial pathogen, varying exposure to host and antibiotics to tease apart the contributions of these selective pressures on pathogen adaptation. After 12 passages, S. aureus evolving in Caenorhabditis elegans nematodes exposed to a sub-minimum inhibitory concentration of antibiotic (oxacillin) became highly virulent, regardless of whether the ancestral pathogen was methicillin-resistant (MRSA) or methicillin-sensitive (MSSA). Host and antibiotic exposure selected for reduced drug susceptibility in MSSA lineages while increasing MRSA total growth outside hosts. We identified mutations in genes involved in complex regulatory networks linking virulence and metabolism, including codY , agr , and gdpP , suggesting that rapid adaptation to infect hosts may have pleiotropic effects. In particular, MSSA populations under selection from host and antibiotic accumulated mutations in the global regulator gene codY , which controls biofilm formation in S. aureus. These populations had indeed evolved more robust biofilms-a trait linked to both virulence and antibiotic resistance-suggesting evolution of one trait can confer multiple adaptive benefits. Despite evolving in similar environments, MRSA and MSSA populations proceeded on divergent evolutionary paths, with MSSA populations exhibiting more similarities across replicate populations. Our results underscore the importance of considering multiple and concurrent selective pressures as drivers of pervasive pathogen traits.
Collapse
|
13
|
Liu L, Wang L, Liu X, Wang B, Guo X, Wang Y, Xu Y, Guan J, Zhao Y. Elucidating the potential of isorhapontigenin in targeting the MgrA regulatory network: a paradigm shift for attenuating MRSA virulence. Antimicrob Agents Chemother 2024; 68:e0061124. [PMID: 39046236 PMCID: PMC11373206 DOI: 10.1128/aac.00611-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/06/2024] [Indexed: 07/25/2024] Open
Abstract
As methicillin-resistant Staphylococcus aureus (MRSA) exhibits formidable resistance to many drugs, the imperative for alternative therapeutic strategies becomes increasingly evident. At the heart of our study is the identification of a novel inhibitor through fluorescence anisotropy assays, specifically targeting the crucial multiple gene regulator A (MgrA) regulatory network in S. aureus. Isorhapontigenin (Iso), a natural compound, exhibits outstanding inhibitory efficacy, modulating bacterial virulence pathways without exerting direct bactericidal activity. This suggests a paradigm shift toward attenuating virulence instead of purely focusing on bacterial elimination. Through comprehensive in vitro and in vivo evaluations, we elucidated the complex interplay between Iso and MgrA, leading to reduced S. aureus adhesion, and overall virulence. At the cellular level, Iso offers significant protection to A549 cells infected with S. aureus, reducing cellular damage. Importantly, Iso augments the chemotaxis of neutrophils, curtailing the immune evasion capabilities of S. aureus. Furthermore, in vivo investigations highlight the notable effectiveness of Iso against MRSA-induced pneumonia and within the Galleria mellonella infection model, underscoring its pivotal role in the evolving realm of antibacterial drug discovery. Significantly, when Iso is used in combination with vancomycin, it outperforms its solo application, indicating a more pronounced therapeutic impact. This seminal research emphasizes Iso's potential as a primary defense against the surge of multidrug-resistant pathogens, heralding new prospects in antimicrobial therapy.
Collapse
Affiliation(s)
- Lihan Liu
- Department of Infectious Diseases and Center of Infectious Diseases and Pathogen Biology, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Jilin University, Changchun, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Li Wang
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, China
| | - Xiaolei Liu
- Department of Infectious Diseases and Center of Infectious Diseases and Pathogen Biology, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Jilin University, Changchun, China
| | - Bingmei Wang
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, China
| | - Xuerui Guo
- School of Pharmaceutical Science, Jilin University, Changchun, China
| | - Yueying Wang
- Department of Orthopedics, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Yueshan Xu
- Department of Orthopedics, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Jiyu Guan
- Department of Infectious Diseases and Center of Infectious Diseases and Pathogen Biology, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Jilin University, Changchun, China
| | - Yicheng Zhao
- Department of Infectious Diseases and Center of Infectious Diseases and Pathogen Biology, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Jilin University, Changchun, China
- China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
14
|
Shil A, Banerjee A, Roy J, Pal M, Das D, Paul R, Maji BK, Sikdar M. The potential antibacterial effects of tea polyphenols. Drug Metab Pers Ther 2024; 39:103-114. [PMID: 39263725 DOI: 10.1515/dmpt-2024-0058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 08/07/2024] [Indexed: 09/13/2024]
Abstract
The current review of tea and its parts is focused on the antibacterial properties, considering the possible applications and modes of action against bacterial illnesses. It shows the backdrop of antibiotic resistance and the huge demand for antibacterial treatments out there. From the interactions with bacterial components, the theory presented that tea polyphenols are antibacterial and therefore would be a substitute or supplementary therapy to the usual antibiotics. The study highlighted the role of tea polyphenols as potential antibacterial compounds that may interact with various bacterial components and different polyphenolic compounds occurring in tea. Future research directions may be directed toward testing more plant-based sources for antibacterial properties, in vivo validation of the studies, and possible synergistic effects with classical antibiotics. By addressing the controversies and disagreements involved, the present understanding of the topic of tea's antibacterial properties and enable the entry of new ways for fighting microorganisms resistant to antibiotics. In conclusion, this review adds to the growing body of evidence regarding the antimicrobial properties of tea and emphasizes the need for further studies that will allow the full exploitation of its therapeutic potential for countering the rising problem of antibiotic resistance in healthcare.
Collapse
Affiliation(s)
- Aparna Shil
- Department of Physiology, Harimohan Ghose College, Kolkata, West Bengal, India
| | - Arnab Banerjee
- Department of Physiology (UG & PG), Serampore College, Hooghly, West Bengal, India
| | - Jayati Roy
- Department of Physiology (UG & PG), Serampore College, Hooghly, West Bengal, India
| | - Manisha Pal
- Department of Physiology (UG & PG), Serampore College, Hooghly, West Bengal, India
| | - Debasmita Das
- Department of Physiology (UG & PG), Serampore College, Hooghly, West Bengal, India
| | - Rajarshi Paul
- Department of Physiology (UG & PG), Serampore College, Hooghly, West Bengal, India
| | - Bithin Kumar Maji
- Department of Physiology (UG & PG), Serampore College, Hooghly, West Bengal, India
| | - Mausumi Sikdar
- Department of Life Sciences, Microbiology, Nutrition and Dietetics Laboratory, Physiology Unit, Presidency University, Kolkata, West Bengal, India
| |
Collapse
|
15
|
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
|
16
|
Ahsan R, Kifayat S, Pooniya KK, Kularia S, Adimalla BS, Sanapalli BKR, Sanapalli V, Sigalapalli DK. Bacterial Histidine Kinase and the Development of Its Inhibitors in the 21st Century. Antibiotics (Basel) 2024; 13:576. [PMID: 39061258 PMCID: PMC11274179 DOI: 10.3390/antibiotics13070576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Bacterial histidine kinase (BHK) is a constituent of the two-component signaling (TCS) pathway, which is responsible for the regulation of a number of processes connected to bacterial pathogenicity, virulence, biofilm development, antibiotic resistance, and bacterial persistence. As BHK regulation is diverse, inhibitors can be developed, such as antibiotic synergists, bacteriostatic/bactericidal agents, virulence inhibitors, and biofilm inhibitors. Inhibition of essential BHK has always been an amenable strategy due to the conserved binding sites of the domains across bacterial species and growth dependence. Hence, an inhibitor of BHK might block multiple TCS regulatory networks. This review describes the TCS system and the role of BHK in bacterial virulence and discusses the available inhibitors of BHK, which is a specific response regulator with essential structural features.
Collapse
Affiliation(s)
- Ragib Ahsan
- Department of Pharmacy, NIMS Institute of Pharmacy, NIMS University, Jaipur 303121, Rajasthan, India (S.K.)
| | - Sumaiya Kifayat
- Department of Pharmacy, NIMS Institute of Pharmacy, NIMS University, Jaipur 303121, Rajasthan, India (S.K.)
| | - Krishan Kumar Pooniya
- Department of Pharmacy, NIMS Institute of Pharmacy, NIMS University, Jaipur 303121, Rajasthan, India (S.K.)
| | - Sunita Kularia
- Department of Pharmacology, NIMS Institute of Pharmacy, NIMS University, Jaipur 303121, Rajasthan, India;
| | - Bhavani Sailu Adimalla
- Department of Pharmaceutical Analysis, Vignan Pharmacy College, Jawaharlal Nehru Technological University, Vadlamudi, Guntur 522213, Andhra Pradesh, India;
| | - Bharat Kumar Reddy Sanapalli
- Department of Pharmacology, School of Pharmacy & Technology Management, SVKM’s Narsee Monjee Institute of Management Studies (NMIMS) Deemed to-be-University, Jadcherla 509301, Hyderabad, India;
| | - Vidyasrilekha Sanapalli
- Department of Pharmaceutical Chemistry, School of Pharmacy & Technology Management, SVKM’s Narsee Monjee Institute of Management Studies (NMIMS) Deemed to-be-University, Jadcherla 509301, Hyderabad, India
| | | |
Collapse
|
17
|
Lee J, Choi JH, Lee J, Cho E, Lee YJ, Lee HS, Oh KB. Halenaquinol Blocks Staphylococcal Protein A Anchoring on Cell Wall Surface by Inhibiting Sortase A in Staphylococcus aureus. Mar Drugs 2024; 22:266. [PMID: 38921577 PMCID: PMC11204543 DOI: 10.3390/md22060266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/05/2024] [Accepted: 06/08/2024] [Indexed: 06/27/2024] Open
Abstract
Sortase A (SrtA) is a cysteine transpeptidase that binds to the periplasmic membrane and plays a crucial role in attaching surface proteins, including staphylococcal protein A (SpA), to the peptidoglycan cell wall. Six pentacyclic polyketides (1-6) were isolated from the marine sponge Xestospongia sp., and their structures were elucidated using spectroscopic techniques and by comparing them to previously reported data. Among them, halenaquinol (2) was found to be the most potent SrtA inhibitor, with an IC50 of 13.94 μM (4.66 μg/mL). Semi-quantitative reverse transcription PCR data suggest that halenaquinol does not inhibit the transcription of srtA and spA, while Western blot analysis and immunofluorescence microscopy images suggest that it blocks the cell wall surface anchoring of SpA by inhibiting the activity of SrtA. The onset and magnitude of the inhibition of SpA anchoring on the cell wall surface in S. aureus that has been treated with halenaquinol at a value 8× that of the IC50 of SrtA are comparable to those for an srtA-deletion mutant. These findings contribute to the understanding of the mechanism by which marine-derived pentacyclic polyketides inhibit SrtA, highlighting their potential as anti-infective agents targeting S. aureus virulence.
Collapse
Affiliation(s)
- Jaepil Lee
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences and Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea; (J.L.); (J.L.); (E.C.)
| | - Jae-Hyeong Choi
- Marine Natural Products Chemistry Laboratory, Korea Institute of Ocean Science and Technology, Busan 49111, Republic of Korea; (J.-H.C.); (Y.-J.L.)
- Department of Applied Ocean Science, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Jayho Lee
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences and Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea; (J.L.); (J.L.); (E.C.)
| | - Eunji Cho
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences and Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea; (J.L.); (J.L.); (E.C.)
| | - Yeon-Ju Lee
- Marine Natural Products Chemistry Laboratory, Korea Institute of Ocean Science and Technology, Busan 49111, Republic of Korea; (J.-H.C.); (Y.-J.L.)
- Department of Applied Ocean Science, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Hyi-Seung Lee
- Marine Natural Products Chemistry Laboratory, Korea Institute of Ocean Science and Technology, Busan 49111, Republic of Korea; (J.-H.C.); (Y.-J.L.)
- Department of Applied Ocean Science, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Ki-Bong Oh
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences and Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea; (J.L.); (J.L.); (E.C.)
| |
Collapse
|
18
|
Sosa-Fajardo A, Díaz-Muñoz C, Van der Veken D, Pradal I, Verce M, Weckx S, Leroy F. Genomic exploration of the fermented meat isolate Staphylococcus shinii IMDO-S216 with a focus on competitiveness-enhancing secondary metabolites. BMC Genomics 2024; 25:575. [PMID: 38849728 PMCID: PMC11161930 DOI: 10.1186/s12864-024-10490-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 06/03/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Staphylococcus shinii appears as an umbrella species encompassing several strains of Staphylococcus pseudoxylosus and Staphylococcus xylosus. Given its phylogenetic closeness to S. xylosus, S. shinii can be found in similar ecological niches, including the microbiota of fermented meats where the species may contribute to colour and flavour development. In addition to these conventional functionalities, a biopreservation potential based on the production of antagonistic compounds may be available. Such potential, however, remains largely unexplored in contrast to the large body of research that is available on the biopreservative properties of lactic acid bacteria. The present study outlines the exploration of the genetic basis of competitiveness and antimicrobial activity of a fermented meat isolate, S. shinii IMDO-S216. To this end, its genome was sequenced, de novo assembled, and annotated. RESULTS The genome contained a single circular chromosome and eight plasmid replicons. Focus of the genomic exploration was on secondary metabolite biosynthetic gene clusters coding for ribosomally synthesized and posttranslationally modified peptides. One complete cluster was coding for a bacteriocin, namely lactococcin 972; the genes coding for the pre-bacteriocin, the ATP-binding cassette transporter, and the immunity protein were also identified. Five other complete clusters were identified, possibly functioning as competitiveness factors. These clusters were found to be involved in various responses such as membrane fluidity, iron intake from the medium, a quorum sensing system, and decreased sensitivity to antimicrobial peptides and competing microorganisms. The presence of these clusters was equally studied among a selection of multiple Staphylococcus species to assess their prevalence in closely-related organisms. CONCLUSIONS Such factors possibly translate in an improved adaptation and competitiveness of S. shinii IMDO-S216 which are, in turn, likely to improve its fitness in a fermented meat matrix.
Collapse
Affiliation(s)
- Ana Sosa-Fajardo
- Research Group of Industrial Microbiology and Food Biotechnology (IMDO), Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cristian Díaz-Muñoz
- Research Group of Industrial Microbiology and Food Biotechnology (IMDO), Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - David Van der Veken
- Research Group of Industrial Microbiology and Food Biotechnology (IMDO), Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Inés Pradal
- Research Group of Industrial Microbiology and Food Biotechnology (IMDO), Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marko Verce
- Research Group of Industrial Microbiology and Food Biotechnology (IMDO), Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Stefan Weckx
- Research Group of Industrial Microbiology and Food Biotechnology (IMDO), Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Frédéric Leroy
- Research Group of Industrial Microbiology and Food Biotechnology (IMDO), Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
19
|
Kerro Dego O, Vidlund J. Staphylococcal mastitis in dairy cows. Front Vet Sci 2024; 11:1356259. [PMID: 38863450 PMCID: PMC11165426 DOI: 10.3389/fvets.2024.1356259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/06/2024] [Indexed: 06/13/2024] Open
Abstract
Bovine mastitis is one of the most common diseases of dairy cattle. Even though different infectious microorganisms and mechanical injury can cause mastitis, bacteria are the most common cause of mastitis in dairy cows. Staphylococci, streptococci, and coliforms are the most frequently diagnosed etiological agents of mastitis in dairy cows. Staphylococci that cause mastitis are broadly divided into Staphylococcus aureus and non-aureus staphylococci (NAS). NAS is mainly comprised of coagulase-negative Staphylococcus species (CNS) and some coagulase-positive and coagulase-variable staphylococci. Current staphylococcal mastitis control measures are ineffective, and dependence on antimicrobial drugs is not sustainable because of the low cure rate with antimicrobial treatment and the development of resistance. Non-antimicrobial effective and sustainable control tools are critically needed. This review describes the current status of S. aureus and NAS mastitis in dairy cows and flags areas of knowledge gaps.
Collapse
Affiliation(s)
- Oudessa Kerro Dego
- Department of Animal Science, University of Tennessee, Knoxville, TN, United States
| | - Jessica Vidlund
- Department of Animal Science, University of Tennessee, Knoxville, TN, United States
- East Tennessee AgResearch and Education Center-Little River Animal and Environmental Unit, University of Tennessee, Walland, TN, United States
| |
Collapse
|
20
|
Podkowik M, Perault AI, Putzel G, Pountain A, Kim J, DuMont AL, Zwack EE, Ulrich RJ, Karagounis TK, Zhou C, Haag AF, Shenderovich J, Wasserman GA, Kwon J, Chen J, Richardson AR, Weiser JN, Nowosad CR, Lun DS, Parker D, Pironti A, Zhao X, Drlica K, Yanai I, Torres VJ, Shopsin B. Quorum-sensing agr system of Staphylococcus aureus primes gene expression for protection from lethal oxidative stress. eLife 2024; 12:RP89098. [PMID: 38687677 PMCID: PMC11060713 DOI: 10.7554/elife.89098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024] Open
Abstract
The agr quorum-sensing system links Staphylococcus aureus metabolism to virulence, in part by increasing bacterial survival during exposure to lethal concentrations of H2O2, a crucial host defense against S. aureus. We now report that protection by agr surprisingly extends beyond post-exponential growth to the exit from stationary phase when the agr system is no longer turned on. Thus, agr can be considered a constitutive protective factor. Deletion of agr resulted in decreased ATP levels and growth, despite increased rates of respiration or fermentation at appropriate oxygen tensions, suggesting that Δagr cells undergo a shift towards a hyperactive metabolic state in response to diminished metabolic efficiency. As expected from increased respiratory gene expression, reactive oxygen species (ROS) accumulated more in the agr mutant than in wild-type cells, thereby explaining elevated susceptibility of Δagr strains to lethal H2O2 doses. Increased survival of wild-type agr cells during H2O2 exposure required sodA, which detoxifies superoxide. Additionally, pretreatment of S. aureus with respiration-reducing menadione protected Δagr cells from killing by H2O2. Thus, genetic deletion and pharmacologic experiments indicate that agr helps control endogenous ROS, thereby providing resilience against exogenous ROS. The long-lived 'memory' of agr-mediated protection, which is uncoupled from agr activation kinetics, increased hematogenous dissemination to certain tissues during sepsis in ROS-producing, wild-type mice but not ROS-deficient (Cybb-/-) mice. These results demonstrate the importance of protection that anticipates impending ROS-mediated immune attack. The ubiquity of quorum sensing suggests that it protects many bacterial species from oxidative damage.
Collapse
Affiliation(s)
- Magdalena Podkowik
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of MedicineNew YorkUnited States
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
| | - Andrew I Perault
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
- Department of Microbiology, NYU Grossman School of MedicineNew YorkUnited States
| | - Gregory Putzel
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
- Department of Microbiology, NYU Grossman School of MedicineNew YorkUnited States
- Microbial Computational Genomic Core Lab, NYU Grossman School of MedicineNew YorkUnited States
| | - Andrew Pountain
- Institute for Systems Genetics; NYU Grossman School of MedicineNew YorkUnited States
| | - Jisun Kim
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical SchoolNewarkUnited States
| | - Ashley L DuMont
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of MedicineNew YorkUnited States
| | - Erin E Zwack
- Department of Microbiology, NYU Grossman School of MedicineNew YorkUnited States
| | - Robert J Ulrich
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of MedicineNew YorkUnited States
| | - Theodora K Karagounis
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
- Ronald O. Perelman Department of Dermatology; NYU Grossman School of MedicineNew YorkUnited States
| | - Chunyi Zhou
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of MedicineNew YorkUnited States
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
| | - Andreas F Haag
- School of Medicine, University of St AndrewsSt AndrewsUnited Kingdom
| | - Julia Shenderovich
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
- Department of Microbiology, NYU Grossman School of MedicineNew YorkUnited States
| | - Gregory A Wasserman
- Department of Surgery, Northwell Health Lenox Hill HospitalNew YorkUnited States
| | - Junbeom Kwon
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of MedicineNew YorkUnited States
| | - John Chen
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
| | - Anthony R Richardson
- Department of Microbiology and Molecular Genetics, University of PittsburghPittsburghUnited States
| | - Jeffrey N Weiser
- Department of Microbiology, NYU Grossman School of MedicineNew YorkUnited States
| | - Carla R Nowosad
- Department of Pathology, NYU Grossman School of MedicineNew YorkUnited States
| | - Desmond S Lun
- Center for Computational and Integrative Biology and Department of Computer Science, Rutgers UniversityCamdenUnited States
| | - Dane Parker
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical SchoolNewarkUnited States
| | - Alejandro Pironti
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
- Department of Microbiology, NYU Grossman School of MedicineNew YorkUnited States
- Microbial Computational Genomic Core Lab, NYU Grossman School of MedicineNew YorkUnited States
| | - Xilin Zhao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen UniversityXiamenChina
| | - Karl Drlica
- Public Health Research Institute, New Jersey Medical School, Rutgers UniversityNew YprkUnited States
- Department of Microbiology, Biochemistry & Molecular Genetics, New Jersey Medical School, Rutgers UniversityNewarkUnited States
| | - Itai Yanai
- Institute for Systems Genetics; NYU Grossman School of MedicineNew YorkUnited States
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of MedicineNew YorkUnited States
| | - Victor J Torres
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
- Department of Microbiology, NYU Grossman School of MedicineNew YorkUnited States
| | - Bo Shopsin
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of MedicineNew YorkUnited States
- Antimicrobial-Resistant Pathogens Program, New York University School of MedicineNew YorkUnited States
- Department of Microbiology, NYU Grossman School of MedicineNew YorkUnited States
| |
Collapse
|
21
|
Yamaguchi J, Manome T, Hara Y, Yamazaki Y, Nakamura Y, Ishibashi M, Takaya A. Physalin H, physalin B, and isophysalin B suppress the quorum-sensing function of Staphylococcus aureus by binding to AgrA. Front Pharmacol 2024; 15:1365815. [PMID: 38659576 PMCID: PMC11039898 DOI: 10.3389/fphar.2024.1365815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
The virulence of Staphylococcus aureus, including methicillin-resistant S. aureus (MRSA), depends on the expression of toxins and virulence factors controlled by the quorum-sensing (QS) system, encoded on the virulence accessory gene regulator (agr) locus. The aim of this study was to identify a phytochemical that inhibits Agr-QS function and to elucidate its mechanism. We screened 577 compounds and identified physalin H, physalin B, and isophysalin B--phytochemicals belonging to physalins found in plants of the Solanaceae family--as novel Agr-QS modulators. Biological analyses and in vitro protein-DNA binding assays suggested that these physalins suppress gene expression related to the Agr-QS system by inhibiting binding of the key response regulator AgrA to the agr promoters, reducing the function of hemolytic toxins downstream of these genes in MRSA. Furthermore, although physalin F suppressed gene expression in the Agr-QS system, its anti-hemolytic activity was lower than that of physalins H, B, and isophysalin B. Conversely, five physalins isolated from the same plant with the ability to suppress Agr-QS did not reduce bacterial Agr-QS activity but inhibited AgrA binding to DNA in vitro. A docking simulation revealed that physalin interacts with the DNA-binding site of AgrA in three docking states. The carbonyl oxygens at C-1 and C-18 of physalins, which can suppress Agr-QS, were directed to residues N201 and R198 of AgrA, respectively, whereas these carbonyl oxygens of physalins, without Agr-QS suppression activity, were oriented in different directions. Next, 100-ns molecular dynamics simulations revealed that the hydrogen bond formed between the carbonyl oxygen at C-15 of physalins and L186 of AgrA functions as an anchor, sustaining the interaction between the carbonyl oxygen at C-1 of physalins and N201 of AgrA. Thus, these results suggest that physalin H, physalin B, and isophysalin B inhibit the interaction of AgrA with the agr promoters by binding to the DNA-binding site of AgrA, suppressing the Agr-QS function of S. aureus. Physalins that suppress the Agr-QS function are proposed as potential lead compounds in the anti-virulence strategy for MRSA infections.
Collapse
Affiliation(s)
- Junpei Yamaguchi
- Department of Infection Control Science, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Teruhisa Manome
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Laboratory of Natural Products Chemistry, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yasumasa Hara
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Faculty of Agriculture, Kagawa University, Takamatsu, Japan
| | - Yuriko Yamazaki
- Cutaneous Allergy and Host Defense, Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Dermatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuumi Nakamura
- Cutaneous Allergy and Host Defense, Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Dermatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masami Ishibashi
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
- School of Pharmacy at Fukuoka, International University of Health and Welfare, Okawa, Japan
| | - Akiko Takaya
- Department of Infection Control Science, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Medical Mycology Research Center, Chiba University, Chiba, Japan
- Plant Molecular Science Center, Chiba University, Chiba, Japan
| |
Collapse
|
22
|
Podkowik M, Perault AI, Putzel G, Pountain A, Kim J, Dumont A, Zwack E, Ulrich RJ, Karagounis TK, Zhou C, Haag AF, Shenderovich J, Wasserman GA, Kwon J, Chen J, Richardson AR, Weiser JN, Nowosad CR, Lun DS, Parker D, Pironti A, Zhao X, Drlica K, Yanai I, Torres VJ, Shopsin B. Quorum-sensing agr system of Staphylococcus aureus primes gene expression for protection from lethal oxidative stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.08.544038. [PMID: 37333372 PMCID: PMC10274873 DOI: 10.1101/2023.06.08.544038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The agr quorum-sensing system links Staphylococcus aureus metabolism to virulence, in part by increasing bacterial survival during exposure to lethal concentrations of H2O2, a crucial host defense against S. aureus. We now report that protection by agr surprisingly extends beyond post-exponential growth to the exit from stationary phase when the agr system is no longer turned on. Thus, agr can be considered a constitutive protective factor. Deletion of agr increased both respiration and fermentation but decreased ATP levels and growth, suggesting that Δagr cells assume a hyperactive metabolic state in response to reduced metabolic efficiency. As expected from increased respiratory gene expression, reactive oxygen species (ROS) accumulated more in the agr mutant than in wild-type cells, thereby explaining elevated susceptibility of Δagr strains to lethal H2O2 doses. Increased survival of wild-type agr cells during H2O2 exposure required sodA, which detoxifies superoxide. Additionally, pretreatment of S. aureus with respiration-reducing menadione protected Δagr cells from killing by H2O2. Thus, genetic deletion and pharmacologic experiments indicate that agr helps control endogenous ROS, thereby providing resilience against exogenous ROS. The long-lived "memory" of agr-mediated protection, which is uncoupled from agr activation kinetics, increased hematogenous dissemination to certain tissues during sepsis in ROS-producing, wild-type mice but not ROS-deficient (Nox2-/-) mice. These results demonstrate the importance of protection that anticipates impending ROS-mediated immune attack. The ubiquity of quorum sensing suggests that it protects many bacterial species from oxidative damage.
Collapse
Affiliation(s)
- Magdalena Podkowik
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of Medicine, New York, NY, USA
- Antimicrobial-Resistant Pathogens Program, New York University School of Medicine, New York, NY, USA
| | - Andrew I. Perault
- Antimicrobial-Resistant Pathogens Program, New York University School of Medicine, New York, NY, USA
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Gregory Putzel
- Antimicrobial-Resistant Pathogens Program, New York University School of Medicine, New York, NY, USA
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, USA
- Microbial Computational Genomic Core Lab, NYU Grossman School of Medicine, New York, NY, USA
| | - Andrew Pountain
- Institute for Systems Genetics; NYU Grossman School of Medicine, New York, NY, USA
| | - Jisun Kim
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School Cancer Center, Newark, NJ, USA
| | - Ashley Dumont
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Erin Zwack
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Robert J. Ulrich
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of Medicine, New York, NY, USA
| | - Theodora K. Karagounis
- Antimicrobial-Resistant Pathogens Program, New York University School of Medicine, New York, NY, USA
- Ronald O. Perelman Department of Dermatology; NYU Grossman School of Medicine, New York, NY, USA
| | - Chunyi Zhou
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of Medicine, New York, NY, USA
- Antimicrobial-Resistant Pathogens Program, New York University School of Medicine, New York, NY, USA
| | - Andreas F. Haag
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Julia Shenderovich
- Antimicrobial-Resistant Pathogens Program, New York University School of Medicine, New York, NY, USA
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Junbeom Kwon
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of Medicine, New York, NY, USA
| | - John Chen
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Anthony R. Richardson
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeffrey N. Weiser
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Carla R. Nowosad
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Desmond S. Lun
- Center for Computational and Integrative Biology and Department of Computer Science, Rutgers University, Camden, NJ, USA
| | - Dane Parker
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers New Jersey Medical School Cancer Center, Newark, NJ, USA
| | - Alejandro Pironti
- Antimicrobial-Resistant Pathogens Program, New York University School of Medicine, New York, NY, USA
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, USA
- Microbial Computational Genomic Core Lab, NYU Grossman School of Medicine, New York, NY, USA
| | - Xilin Zhao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian Province, China
| | - Karl Drlica
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, USA
- Department of Microbiology, Biochemistry & Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Itai Yanai
- Institute for Systems Genetics; NYU Grossman School of Medicine, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA
| | - Victor J. Torres
- Antimicrobial-Resistant Pathogens Program, New York University School of Medicine, New York, NY, USA
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Bo Shopsin
- Department of Medicine, Division of Infectious Diseases, NYU Grossman School of Medicine, New York, NY, USA
- Antimicrobial-Resistant Pathogens Program, New York University School of Medicine, New York, NY, USA
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
23
|
Dagnaw M, Bazezew M, Mengistu B, Anagaw B, Mebratu AS. Rate of Beta-Lactam Resistance and Epidemiological Features of S. Aureus-Associated Bovine Mastitis in Cross-Bred Ethiopian Cows: Systematic Review. VETERINARY MEDICINE (AUCKLAND, N.Z.) 2024; 15:39-55. [PMID: 38433734 PMCID: PMC10908337 DOI: 10.2147/vmrr.s415339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 02/08/2024] [Indexed: 03/05/2024]
Abstract
Background Dairy cows get mastitis from a common infection called Staphylococcus aureus. Because of its broad distribution across diverse populations and capacity to acquire antibiotic resistance, this particular bacterial strain presents a serious threat to public health. The main goals of this study were to determine the beta-lactam resistance profile of S. aureus in Ethiopian dairy cows and to offer thorough epidemiological data. Methods We employed manual searches, Web of Science, PubMed Central, and Google Scholar HINARI for electronic bibliographic data. Results Twenty-six epidemiological studies were included in this systematic review. Of these studies, 12 articles in Oromia, 4 articles in Addis Ababa, 4 articles in Southern Nations, Nationalities, and People's (SNNPRS), 3 articles in Tigray, and 3 articles in Amhara region. The average prevalence S. aureus were 34.3% in Oromia, 40.2% in Amhara, 39.5 in AA, 40% in Tigray and 21% in SNNPRS. The antimicrobial resistance rate of S. aureus, specifically in relation to beta-lactam drugs, exhibited an average estimation. Notably, penicillin resistance reached a rate of 75%, while amoxicillin resistance stood at 67%. Furthermore, it was determined that, when treating S. aureus, the resistance rates to ampicillin and cephalosporin were 50% and 57%, respectively. Conclusion The results of this analysis have demonstrated a considerable rise in S. aureus prevalence and beta-lactam resistance within the Ethiopian geographic environment. This emphasizes the critical need for alternate therapeutic approaches and preventative measures in order to successfully lessen the disease's extensive spread and detrimental effects across the nation.
Collapse
Affiliation(s)
- Melkie Dagnaw
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine and Animal Science, University of Gondar, Gondar, Ethiopia
| | - Marshet Bazezew
- Department of Epidemiology, College of Veterinary Medicine and Animal Science, University of Gondar, Gondar, Ethiopia
| | - Bemrew Mengistu
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Science, University of Gondar, Gondar, Ethiopia
| | - Birhan Anagaw
- Department of Pathology, College of Veterinary Medicine and Animal Science, University of Gondar, Gondar, Ethiopia
| | - Atsede Solomon Mebratu
- Department of Pharmacy, College of Veterinary Medicine and Animal Science, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
24
|
Savin A, Anderson EE, Dyzenhaus S, Podkowik M, Shopsin B, Pironti A, Torres VJ. Staphylococcus aureus senses human neutrophils via PerR to coordinate the expression of the toxin LukAB. Infect Immun 2024; 92:e0052623. [PMID: 38235972 PMCID: PMC10863418 DOI: 10.1128/iai.00526-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/19/2024] Open
Abstract
Staphylococcus aureus is a gram-positive pathogen that poses a major health concern, in part due to its large array of virulence factors that allow infection and evasion of the immune system. One of these virulence factors is the bicomponent pore-forming leukocidin LukAB. The regulation of lukAB expression is not completely understood, especially in the presence of immune cells such as human polymorphonuclear neutrophils (hPMNs). Here, we screened for transcriptional regulators of lukAB during the infection of primary hPMNs. We uncovered that PerR, a peroxide sensor, is vital for hPMN-mediated induction of lukAB and that PerR upregulates cytotoxicity during the infection of hPMNs. Exposure of S. aureus to hydrogen peroxide (H2O2) alone also results in increased lukAB promoter activity, a phenotype dependent on PerR. Collectively, our data suggest that S. aureus uses PerR to sense the H2O2 produced by hPMNs to stimulate the expression of lukAB, allowing the bacteria to withstand these critical innate immune cells.IMPORTANCEStaphylococcus aureus utilizes a diverse set of virulence factors, such as leukocidins, to subvert human neutrophils, but how these toxins are regulated is incompletely defined. Here, we identified the peroxide-sensitive repressor, PerR, as a required protein involved in the induction of lukAB in the presence of primary human neutrophils, a phenotype directly linked to the ability of PerR to sense H2O2. Thus, we show that S. aureus coordinates sensing and resistance to oxidative stress with toxin production to promote pathogen survival.
Collapse
Affiliation(s)
- Avital Savin
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Department of Biology, New York University, New York, New York, USA
| | - Exene E. Anderson
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Sophie Dyzenhaus
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Magdalena Podkowik
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
- Division of Infectious Diseases, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Bo Shopsin
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
- Division of Infectious Diseases, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Alejandro Pironti
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Department of Host-Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
25
|
Vinodhini V, Kavitha M. Deciphering agr quorum sensing in Staphylococcus aureus: insights and therapeutic prospects. Mol Biol Rep 2024; 51:155. [PMID: 38252331 DOI: 10.1007/s11033-023-08930-3] [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: 08/01/2023] [Accepted: 10/10/2023] [Indexed: 01/23/2024]
Abstract
The emergence of superbugs like methicillin-resistant Staphylococcus aureus exposed the limitations of treating microbial infections using antibiotics. At present, the discovery of novel and convincing therapeutic methods are being executed increasingly as possible substitutes to conventional antibiotic therapies. The quorum sensing helps Staphylococcus aureus become more viable through their signaling mechanisms. In recent years, targeting the prominent factors of quorum sensing has obtained remarkable attention as a futuristic approach to dealing with bacterial pathogenicity. The standard antibiotic therapy intends to inhibit the organism by targeting specific molecules and afford a chance for the evolution of antibiotic resistance. This prompts the development of novel therapeutic strategies like inhibiting quorum sensing that can limit bacterial virulence by decreasing the selective pressure, thereby restricting antibiotic resistance evolution. This review furnishes new insights into the accessory gene regulator quorum sensing in Staphylococcus aureus and its inhibition by targeting the genes that regulate the operon. Further, this review comprehensively explores the inhibitors reported up to date and their specific targets and discusses their potentially ineffective alternative therapy against methicillin-resistant Staphylococcus aureus.
Collapse
Affiliation(s)
- V Vinodhini
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - M Kavitha
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
26
|
Bhattacharya M, Horswill AR. The role of human extracellular matrix proteins in defining Staphylococcus aureus biofilm infections. FEMS Microbiol Rev 2024; 48:fuae002. [PMID: 38337187 PMCID: PMC10873506 DOI: 10.1093/femsre/fuae002] [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: 07/19/2023] [Revised: 01/26/2024] [Accepted: 02/07/2024] [Indexed: 02/12/2024] Open
Abstract
Twenty to forty one percent of the world's population is either transiently or permanently colonized by the Gram-positive bacterium, Staphylococcus aureus. In 2017, the CDC designated methicillin-resistant S. aureus (MRSA) as a serious threat, reporting ∼300 000 cases of MRSA-associated hospitalizations annually, resulting in over 19 000 deaths, surpassing that of HIV in the USA. S. aureus is a proficient biofilm-forming organism that rapidly acquires resistance to antibiotics, most commonly methicillin (MRSA). This review focuses on a large group of (>30) S. aureus adhesins, either surface-associated or secreted that are designed to specifically bind to 15 or more of the proteins that form key components of the human extracellular matrix (hECM). Importantly, this includes hECM proteins that are pivotal to the homeostasis of almost every tissue environment [collagen (skin), proteoglycans (lung), hemoglobin (blood), elastin, laminin, fibrinogen, fibronectin, and fibrin (multiple organs)]. These adhesins offer S. aureus the potential to establish an infection in every sterile tissue niche. These infections often endure repeated immune onslaught, developing into chronic, biofilm-associated conditions that are tolerant to ∼1000 times the clinically prescribed dose of antibiotics. Depending on the infection and the immune response, this allows S. aureus to seamlessly transition from colonizer to pathogen by subtly manipulating the host against itself while providing the time and stealth that it requires to establish and persist as a biofilm. This is a comprehensive discussion of the interaction between S. aureus biofilms and the hECM. We provide particular focus on the role of these interactions in pathogenesis and, consequently, the clinical implications for the prevention and treatment of S. aureus biofilm infections.
Collapse
Affiliation(s)
- Mohini Bhattacharya
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, United States
- Department of Veterans Affairs, Eastern Colorado Health Care System, Aurora, CO 80045, United States
| |
Collapse
|
27
|
Meireles D, Pombinho R, Cabanes D. Signals behind Listeria monocytogenes virulence mechanisms. Gut Microbes 2024; 16:2369564. [PMID: 38979800 PMCID: PMC11236296 DOI: 10.1080/19490976.2024.2369564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/13/2024] [Indexed: 07/10/2024] Open
Abstract
The tight and coordinated regulation of virulence gene expression is crucial to ensure the survival and persistence of bacterial pathogens in different contexts within their hosts. Considering this, bacteria do not express virulence factors homogenously in time and space, either due to their associated fitness cost or to their detrimental effect at specific infection stages. To efficiently infect and persist into their hosts, bacteria have thus to monitor environmental cues or chemical cell-to-cell signaling mechanisms that allow their transition from the external environment to the host, and therefore adjust gene expression levels, intrinsic biological activities, and appropriate behaviors. Listeria monocytogenes (Lm), a major Gram-positive facultative intracellular pathogen, stands out for its adaptability and capacity to thrive in a wide range of environments. Because of that, Lm presents itself as a significant concern in food safety and public health, that can lead to potentially life-threatening infections in humans. A deeper understanding of the intricate bacterial virulence mechanisms and the signals that control them provide valuable insights into the dynamic interplay between Lm and the host. Therefore, this review addresses the role of some crucial signals behind Lm pathogenic virulence mechanisms and explores how the ability to assimilate and interpret these signals is fundamental for pathogenesis, identifying potential targets for innovative antimicrobial strategies.
Collapse
Affiliation(s)
- Diana Meireles
- Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- Group of Molecular Microbiology, IBMC, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar – ICBAS, Porto, Portugal
| | - Rita Pombinho
- Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- Group of Molecular Microbiology, IBMC, Porto, Portugal
| | - Didier Cabanes
- Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- Group of Molecular Microbiology, IBMC, Porto, Portugal
| |
Collapse
|
28
|
Yang P, Yang J, Lin T, Liu Q, Yin Y, Chen D, Yang S. Efficient Genome Editing in Most Staphylococcus aureus by Using the Restriction-Modification System Silent CRISPR-Cas9 Toolkit. ACS Synth Biol 2023; 12:3340-3351. [PMID: 37830328 DOI: 10.1021/acssynbio.3c00339] [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: 10/14/2023]
Abstract
Staphylococcus aureus is a clinically important pathogen that threatens human health due to its strong pathogenicity and drug resistance, leading to meningitis, endocarditis, and skin and soft tissue infections. Genetic manipulation in S. aureus is a powerful approach for characterizing the molecular mechanisms of bacterial drug resistance, pathogenicity, and virulence. However, a strong restriction barrier presents a major obstacle to the extensive utilization of genetic manipulation tools in clinical isolates of S. aureus. Here, we constructed a restriction-modification (RM) system silent CRISPR-Cas9 toolkit that synonymously eliminated the type I RM targets of S. aureus from plasmids, downsized plasmids using minicircle technology, and combined with a plasmid artificial modification (PAM) method to circumvent the type II RM system. The RM-silent CRISPR-Cas9 toolkit enables a significant improvement in transformation (105-106 transformants per microgram plasmid in strains we tested) and high-success efficiency editing for gene deletion (knockout strain obtained in one-round electroporation) in a wide range of S. aureus species including clinical isolates of unknown genetic background. The RM-silent CRISPR-Cas9 toolkits could expedite the process of mutant construction in most S. aureus strains, and this approach could be applied to the design of other genetic toolkit plasmids for utilization in a wider range of S. aureus strains.
Collapse
Affiliation(s)
- Ping Yang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Junjie Yang
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Ting Lin
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qi Liu
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yu Yin
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Daijie Chen
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
- State Key Laboratory of Microbial Metabolism, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Sheng Yang
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| |
Collapse
|
29
|
Refai MY, Elazzazy AM, Desouky SE, Abu-Elghait M, Fayed EA, Alajel SM, Alajlan AA, Albureikan MO, Nakayama J. Interception of Epoxide ring to quorum sensing system in Enterococcus faecalis and Staphylococcus aureus. AMB Express 2023; 13:126. [PMID: 37946062 PMCID: PMC10636001 DOI: 10.1186/s13568-023-01633-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
Quorum sensing inhibitor (QSI) has been attracting attention as anti-virulence agent which disarms pathogens of their virulence rather than killing them. QSI marking cyclic peptide-mediated QS in Gram-positive bacteria is an effective tool to overcome the crisis of antibiotic-dependent chemotherapy due to the emergence of drug resistance strain, e.g., methicillin resistant Staphylococcus aureus (MRSA) and Vancomycin resistant Enterococci (VRE). From a semi-large-scale screening thus far carried out, two Epoxide compounds, Ambuic acid and Synerazol, have been found to efficiently block agr and fsr QS systems, suggesting that the Epoxide group is involved in the mode of action of these QSIs. To address this notion, known natural Epoxide compounds, Cerulenin and Fosfomycin were examined for QSI activity for the agr and fsr systems in addition to in silico and SAR studies. As a result, most of investigated Epoxide containing antibiotics correlatively interfere with QSI activity for the agr and fsr systems under sublethal concentrations.
Collapse
Affiliation(s)
- Mohammed Y Refai
- Department of Biological Science, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Ahmed M Elazzazy
- Department of Biological Science, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Said E Desouky
- Laboratory of Microbial Technology, Division of Systems Bioengineering, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, 819-0395, Fukuoka, Japan
- Department of Botany and Microbiology, Faculty of Science, Al-Azhar University, 11884, Cairo, Egypt
| | - Mohammed Abu-Elghait
- Department of Botany and Microbiology, Faculty of Science, Al-Azhar University, 11884, Cairo, Egypt.
| | - Eman A Fayed
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy (Girls), Al-Azhar University, 11754, Cairo, Egypt
| | - Sulaiman M Alajel
- Reference Laboratory for Microbiology, Executive Department of Reference Laboratories, Research and Laboratories Sector, Saudi Food and Drug Authority (SFDA), Riyadh, Saudi Arabia
| | - Abdullah A Alajlan
- Microbial Identification Division, Reference Laboratory for Microbiology, Executive Department of Reference Laboratories, Research and Laboratories Sector, Saudi Food and Drug Authority (SFDA), Riyadh, Saudi Arabia
| | - Mona O Albureikan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Jiro Nakayama
- Laboratory of Microbial Technology, Division of Systems Bioengineering, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, 819-0395, Fukuoka, Japan
| |
Collapse
|
30
|
Nappi F, Avtaar Singh SS, Jitendra V, Fiore A. Bridging Molecular and Clinical Sciences to Achieve the Best Treatment of Enterococcus faecalis Endocarditis. Microorganisms 2023; 11:2604. [PMID: 37894262 PMCID: PMC10609379 DOI: 10.3390/microorganisms11102604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/14/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Enterococcus faecalis (E. faecalis) is a commensal bacterium that causes various infections in surgical sites, the urinary tract, and blood. The bacterium is becoming a significant concern because it tends to affect the elderly population, which has a high prevalence of undiagnosed degenerative valvular disease and is often subjected to invasive procedures and implanted medical devices. The bacterium's actions are influenced by specific characteristics like pili activity and biofilm formation. This resistance significantly impedes the effectiveness of numerous antibiotic therapies, particularly in cases of endocarditis. While current guidelines recommend antimicrobial therapy, the emergence of resistant strains has introduced complexity in managing these patients, especially with the increasing use of transcatheter therapies for those who are not suitable for surgery. Presentations of the condition are often varied and associated with generalised symptoms, which may pose a diagnostic challenge. We share our encounter with a case study that concerns an octogenarian who had a TAVI valve and developed endocarditis. We also conducted a literature review to identify the essential treatment algorithms for such cases.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
| | | | - Vikram Jitendra
- Department of Cardiothoracic Surgery, Aberdeen Royal Infirmary, Aberdeen AB25 2ZN, UK;
| | - Antonio Fiore
- Department of Cardiac Surgery, Hôpitaux Universitaires Henri Mondor, Assistance Publique-Hôpitaux de Paris, 94000 Creteil, France;
| |
Collapse
|
31
|
Nappi F, Schoell T, Spadaccio C, Acar C, da Costa FDA. A Literature Review on the Use of Aortic Allografts in Modern Cardiac Surgery for the Treatment of Infective Endocarditis: Is There Clear Evidence or Is It Merely a Perception? Life (Basel) 2023; 13:1980. [PMID: 37895362 PMCID: PMC10608498 DOI: 10.3390/life13101980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/18/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Infective valve endocarditis is caused by different pathogens and 60% of those involve the aortic valve with valve failure. Although S. aureus is recognized as the most frequently isolated causative bacterium associated with IE in high-income countries, Gram-positive cocci nevertheless play a crucial role in promoting infection in relation to their adhesive matrix molecules. The presence of pili on the surface of Gram-positive bacteria such as in different strains of Enterococcus faecalis and Streptococcus spp., grants these causative pathogens a great offensive capacity due to the formation of biofilms and resistance to antibiotics. The indications and timing of surgery in endocarditis are debated as well as the choice of the ideal valve substitute to replace the diseased valve(s) when repair is not possible. We reviewed the literature and elaborated a systematic approach to endocarditis management based on clinical, microbiological, and anatomopathological variables known to affect postoperative outcomes with the aim to stratify the patients and orient decision making. From this review emerges significant findings on the risk of infection in the allograft used in patients with endocarditis and no endocarditis etiology suggesting that the use of allografts has proved safety and effectiveness in patients with both pathologies.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France;
| | - Thibaut Schoell
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France;
| | - Cristiano Spadaccio
- Cardiothoracic Surgery, Lancashire Cardiac Center, Blackpool Victoria Hospital, Blackpool FY3 8NP, UK;
| | - Christophe Acar
- Department of Cardiothoracic Surgery, Hôpital Pitié-Salpêtrière, Boulevard de Hôpital 47-83, 75013 Paris, France;
| | - Francisco Diniz Affonso da Costa
- Department of Cardiovascular Surgery, Instituto de Neurologia e Cardiologia de Curitiba—INC Cardio, Curitiba 81210-310, Parana, Brazil;
| |
Collapse
|
32
|
Kim SY, Kim M, Kim TJ. Regulation of σ B-Dependent Biofilm Formation in Staphylococcus aureus through Strain-Specific Signaling Induced by Diosgenin. Microorganisms 2023; 11:2376. [PMID: 37894034 PMCID: PMC10609180 DOI: 10.3390/microorganisms11102376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/15/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
Staphylococcus aureus is a commensal skin bacterium and a causative agent of infectious diseases. Biofilm formation in S. aureus is a mechanism that facilitates the emergence of resistant strains. This study proposes a mechanism for the regulation of biofilm formation in S. aureus through strain-specific physiological changes induced by the plant steroid diosgenin. A comparison of diosgenin-induced changes in the expression of regulatory genes associated with physiological changes revealed the intracellular regulatory mechanisms involved in biofilm formation. Diosgenin reduced biofilm formation in S. aureus ATCC 6538 and methicillin-resistant S. aureus (MRSA) CCARM 3090 by 39% and 61%, respectively. Conversely, it increased biofilm formation in S. aureus ATCC 29213 and MRSA CCARM 3820 by 186% and 582%, respectively. Cell surface hydrophobicity and extracellular protein and carbohydrate contents changed in a strain-specific manner in response to biofilm formation. An assessment of the changes in gene expression associated with biofilm formation revealed that diosgenin treatment decreased the expression of icaA and spa and increased the expression of RNAIII, agrA, sarA, and sigB in S. aureus ATCC 6538 and MRSA CCARM 3090; however, contrasting gene expression changes were noted in S. aureus ATCC 29213 and MRSA CCARM 3820. These results suggest that a regulatory mechanism of biofilm formation is that activated sigB expression sequentially increases the expression of sarA, agrA, and RNAIII. This increased RNAIII expression decreases the expression of spa, a surface-associated adhesion factor. An additional regulatory mechanism of biofilm formation is that activated sigB expression decreases the expression of an unknown regulator that increases the expression of icaA. This in turn decreases the expression of icaA, which decreases the synthesis of polysaccharide intercellular adhesins and ultimately inhibits biofilm formation. By assessing strain-specific contrasting regulatory signals induced by diosgenin in S. aureus without gene mutation, this study elucidated the signal transduction mechanisms that regulate biofilm formation based on physiological and gene expression changes.
Collapse
Affiliation(s)
| | | | - Tae-Jong Kim
- Department of Forest Products and Biotechnology, Kookmin University, Seoul 02707, Republic of Korea
| |
Collapse
|
33
|
Hachem AA, Filkins LM, Kidane YH, Raj P, Tareen NG, Arana CA, Muthukrishnan G, Copley LA. Staphylococcus aureus isolates from children with clinically differentiated osteomyelitis exhibit distinct transcriptomic signatures. PLoS One 2023; 18:e0288758. [PMID: 37561761 PMCID: PMC10414669 DOI: 10.1371/journal.pone.0288758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 07/04/2023] [Indexed: 08/12/2023] Open
Abstract
There is substantial genomic heterogeneity among Staphylococcus aureus isolates of children with acute hematogenous osteomyelitis (AHO) but transcriptional behavior of clinically differentiated strains has not been previously described. This study evaluates transcriptional activity of S. aureus isolates of children with AHO that may regulate metabolism, biosynthesis, or virulence during bacterial growth and pathogenesis. In vitro growth kinetics were compared between three S. aureus clinical isolates from children with AHO who had mild, moderate, and severe illness. Total RNA sequencing was performed for each isolate at six separate time points throughout the logarithmic phase of growth. The NASA RNA-Sequencing Consensus Pipeline was used to identify differentially expressed genes allowing for 54 comparisons between the three isolates during growth. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment pathways were used to evaluate transcriptional variation in metabolism, biosynthesis pathways and virulence potential of the isolates. The S. aureus isolates demonstrated differing growth kinetics under standardized conditions with the mild isolate having higher optical densities with earlier and higher peak rates of growth than that of the other isolates (p<0.001). Enrichment pathway analysis established distinct transcriptional signatures according to both sampling time and clinical severity. Moderate and severe isolates demonstrated pathways of bacterial invasion, S. aureus infection, quorum sensing and two component systems. In comparison, the mild strain favored biosynthesis and metabolism. These findings suggest that transcriptional regulation during the growth of S. aureus may impact the pathogenetic mechanisms involved in the progression of severity of illness in childhood osteomyelitis. The clinical isolates studied demonstrated a tradeoff between growth and virulence. Further investigation is needed to evaluate these transcriptional pathways in an animal model or during active clinical infections of children with AHO.
Collapse
Affiliation(s)
- Ahmad A. Hachem
- Department of Pediatrics, University of Florida College of Medicine –Jacksonville, Jacksonville, FL, United States of America
| | - Laura M. Filkins
- Department of Microbiology, University of Texas Southwestern, Children’s Health System of Texas, Dallas, TX, United States of America
| | - Yared H. Kidane
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, United States of America
| | - Prithvi Raj
- Microbiome Research Laboratory, University of Texas Southwestern, Dallas, TX, United States of America
| | - Naureen G. Tareen
- Department of Pediatric Orthopaedic Surgery, Children’s Health System of Texas, Dallas, TX, United States of America
| | - Carlos A. Arana
- Genomics Core, University of Texas Southwestern, Dallas, TX, United States of America
| | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States of America
| | - Lawson A. Copley
- Department of Pediatric Orthopaedic Surgery, Children’s Health System of Texas, Dallas, TX, United States of America
- Department of Pediatric Orthopaedic Surgery, University of Texas Southwestern, Dallas, TX, United States of America
| |
Collapse
|
34
|
Tran NN, Morrisette T, Jorgensen SCJ, Orench-Benvenutti JM, Kebriaei R. Current therapies and challenges for the treatment of Staphylococcus aureus biofilm-related infections. Pharmacotherapy 2023; 43:816-832. [PMID: 37133439 DOI: 10.1002/phar.2806] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 05/04/2023]
Abstract
Staphylococcus aureus is a major cause of nosocomial and community-acquired infections and contributes to significant increase in morbidity and mortality especially when associated with medical devices and in biofilm form. Biofilm structure provides a pathway for the enrichment of resistant and persistent phenotypes of S. aureus leading to relapse and recurrence of infection. Minimal diffusion of antibiotics inside biofilm structure leads to heterogeneity and distinct physiological activity. Additionally, horizontal gene transfer between cells in proximity adds to the challenges associated with eradication of biofilms. This narrative review focuses on biofilm-associated infections caused by S. aureus, the impact of environmental conditions on biofilm formation, interactions inside biofilm communities, and the clinical challenges that they present. Conclusively, potential solutions, novel treatment strategies, combination therapies, and reported alternatives are discussed.
Collapse
Affiliation(s)
- Nikki N Tran
- Department of Pharmacy, The Ohio State University Wexner Medical Center - The James Cancer Hospital and Solove Research Institute, Columbus, Ohio, USA
| | - Taylor Morrisette
- Department of Clinical Pharmacy and Outcomes Sciences, Medical University of South Carolina College of Pharmacy, Charleston, South Carolina, USA
- Department of Pharmacy Services, Medical University of South Carolina Shawn Jenkins Children's Hospital, Charleston, South Carolina, USA
| | - Sarah C J Jorgensen
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - José M Orench-Benvenutti
- P3 Research Laboratory, Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Razieh Kebriaei
- P3 Research Laboratory, Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
35
|
Jiang F, Chen Y, Yu J, Zhang F, Liu Q, He L, Musha H, Du J, Wang B, Han P, Chen X, Tang J, Li M, Shen H. Repurposed Fenoprofen Targeting SaeR Attenuates Staphylococcus aureus Virulence in Implant-Associated Infections. ACS CENTRAL SCIENCE 2023; 9:1354-1373. [PMID: 37521790 PMCID: PMC10375895 DOI: 10.1021/acscentsci.3c00499] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Indexed: 08/01/2023]
Abstract
Implant-associated infections (IAIs) caused by S. aureus can result in serious challenges after orthopedic surgery. Due to biofilm formation and antibiotic resistance, this refractory infection is highly prevalent, and finding drugs to attenuate bacterial virulence is becoming a rational alternative strategy. In S. aureus, the SaeRS two-component system (TCS) plays a key role in the production of over 20 virulence factors and the pathogenesis of the bacterium. Here, by conducting a structure-based virtual screening against SaeR, we identified that fenoprofen, a USA Food and Drug Administration (FDA)-approved nonsteroid anti-inflammatory drug (NSAID), had excellent inhibitory potency against the response regulator SaeR protein. We showed that fenoprofen attenuated the virulence of S. aureus without drug resistance. In addition, it was helpful in relieving osteolysis and restoring the walking ability of mice in vitro and in implant-associated infection models. More importantly, fenoprofen treatment suppressed biofilm formation and changed the biofilm structure, which caused S. aureus to form loose and porous biofilms that were more vulnerable to infiltration and elimination by leukocytes. Our results reveal that fenoprofen is a potent antivirulence agent with potential value in clinical applications and that SaeR is a drug target against S. aureus implant-associated infections.
Collapse
Affiliation(s)
- Feng Jiang
- Department
of Orthopedics, Shanghai Sixth People’s
Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Yingjia Chen
- Drug
Discovery and Design Center, State Key Laboratory of Drug Research,
Shanghai Institute of Materia Medica, Chinese
Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
- Department
of Pharmacy, University of Chinese Academy
of Sciences, No.19A Yuan
Road, Beijing 100049, China
| | - Jinlong Yu
- Department
of Orthopedics, Shanghai Sixth People’s
Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Feiyang Zhang
- Department
of Orthopedics, Shanghai Sixth People’s
Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Qian Liu
- Department
of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Lei He
- Department
of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Hamushan Musha
- Department
of Orthopedics, Shanghai Sixth People’s
Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Jiafei Du
- Department
of Orthopedics, Shanghai Sixth People’s
Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Boyong Wang
- Department
of Orthopedics, Shanghai Sixth People’s
Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Pei Han
- Department
of Orthopedics, Shanghai Sixth People’s
Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Xiaohua Chen
- Department
of Infectious Diseases, Shanghai Sixth People’s
Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Jin Tang
- Department
of Clinical Laboratory, Shanghai Sixth People’s
Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Min Li
- Department
of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
- Faculty of
Medical Laboratory Science, Shanghai Jiaotong
University School of Medicine, Shanghai 200025, China
| | - Hao Shen
- Department
of Orthopedics, Shanghai Sixth People’s
Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| |
Collapse
|
36
|
Patel H, Rawat S. A genetic regulatory see-saw of biofilm and virulence in MRSA pathogenesis. Front Microbiol 2023; 14:1204428. [PMID: 37434702 PMCID: PMC10332168 DOI: 10.3389/fmicb.2023.1204428] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/30/2023] [Indexed: 07/13/2023] Open
Abstract
Staphylococcus aureus is one of the most common opportunistic human pathogens causing several infectious diseases. Ever since the emergence of the first methicillin-resistant Staphylococcus aureus (MRSA) strain decades back, the organism has been a major cause of hospital-acquired infections (HA-MRSA). The spread of this pathogen across the community led to the emergence of a more virulent subtype of the strain, i.e., Community acquired Methicillin resistant Staphylococcus aureus (CA-MRSA). Hence, WHO has declared Staphylococcus aureus as a high-priority pathogen. MRSA pathogenesis is remarkable because of the ability of this "superbug" to form robust biofilm both in vivo and in vitro by the formation of polysaccharide intercellular adhesin (PIA), extracellular DNA (eDNA), wall teichoic acids (WTAs), and capsule (CP), which are major components that impart stability to a biofilm. On the other hand, secretion of a diverse array of virulence factors such as hemolysins, leukotoxins, enterotoxins, and Protein A regulated by agr and sae two-component systems (TCS) aids in combating host immune response. The up- and downregulation of adhesion genes involved in biofilm formation and genes responsible for synthesizing virulence factors during different stages of infection act as a genetic regulatory see-saw in the pathogenesis of MRSA. This review provides insight into the evolution and pathogenesis of MRSA infections with a focus on genetic regulation of biofilm formation and virulence factors secretion.
Collapse
Affiliation(s)
| | - Seema Rawat
- Microbiology Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar, Gujarat, India
| |
Collapse
|
37
|
Polaske TJ, West KHJ, Zhao K, Widner DL, York JT, Blackwell HE. Chemical and biomolecular insights into the Staphylococcus aureus agr quorum sensing system: Current progress and ongoing challenges. Isr J Chem 2023; 63:e202200096. [PMID: 38765792 PMCID: PMC11101167 DOI: 10.1002/ijch.202200096] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Indexed: 03/19/2023]
Abstract
Staphylococcus aureus is a ubiquitous bacterium that has become a major threat to human health due to its extensive toxin production and tremendous capacity for antibiotic resistance (e.g., MRSA "superbug" infections). Amid a worsening antibiotic resistance crisis, new strategies to combat this deadly microbe that remove the selective pressure of traditional approaches are in high demand. S. aureus utilizes an accessory gene regulator (agr) quorum sensing network to monitor its local cellular population and trigger a devastating communal attack, like an invading horde, once a threshold cell density has been reached. The role of the agr system in a range of disease types is still being unraveled. Herein, we discuss the present-day biochemical understanding of agr along with unresolved details, describe its connection to the progression of infection, and review how chemical strategies have been implemented to study and intercept this signaling pathway. This research is illuminating the potential of agr as an anti-virulence target in S. aureus and should inform the study of similar, yet less studied, agr systems in related bacterial pathogens.
Collapse
Affiliation(s)
- Thomas J. Polaske
- Department of Chemistry, University of Wisconsin–Madison, 1101 University Ave., Madison, WI 53706 USA
| | - Korbin H. J. West
- Department of Chemistry, University of Wisconsin–Madison, 1101 University Ave., Madison, WI 53706 USA
| | - Ke Zhao
- Department of Chemistry, University of Wisconsin–Madison, 1101 University Ave., Madison, WI 53706 USA
| | - Danielle L. Widner
- Department of Chemistry, University of Wisconsin–Madison, 1101 University Ave., Madison, WI 53706 USA
| | - Jordan T. York
- Department of Chemistry, University of Wisconsin–Madison, 1101 University Ave., Madison, WI 53706 USA
| | - Helen E. Blackwell
- Department of Chemistry, University of Wisconsin–Madison, 1101 University Ave., Madison, WI 53706 USA
| |
Collapse
|
38
|
Nurxat N, Wang L, Wang Q, Li S, Jin C, Shi Y, Wulamu A, Zhao N, Wang Y, Wang H, Li M, Liu Q. Commensal Staphylococcus epidermidis Defends against Staphylococcus aureus through SaeRS Two-Component System. ACS OMEGA 2023; 8:17712-17718. [PMID: 37251147 PMCID: PMC10210170 DOI: 10.1021/acsomega.3c00263] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 04/26/2023] [Indexed: 05/31/2023]
Abstract
Staphylococcus aureus is a high-virulent Gram-positive pathogen that is responsible for a serious of diseases. The emergence of antibiotic-resistant S. aureus poses a significant challenge in terms of treatment. The recent research on the human microbiome suggested that the application of commensal bacteria is a new strategy for combating pathogenic infections. Staphylococcus epidermidis, one of the most abundant species in the nasal microbiome, is able to inhibit the colonization of S. aureus. However, during bacterial competition, S. aureus undergoes evolutionary changes to adapt to the diverse environment. Our study has demonstrated that the nasal colonized S. epidermidis possesses the ability to inhibit the hemolytic activity of S. aureus. Moreover, we deciphered another layer of mechanism to inhibit S. aureus colonization by S. epidermidis. The active component present in the cell-free culture of S. epidermidis was found to significantly reduce the hemolytic activity of S. aureus in SaeRS- and Agr-dependent manner. Specifically, the hemolytic inhibition on the S. aureus Agr-I type by S. epidermidis is primarily dependent on the SaeRS two-component system. The active component is characterized as a small molecule that is heat sensitive and protease resistant. Critically, S. epidermidis significantly inhibit the virulence of S. aureus in a mouse skin abscess model, suggesting that the active compound could potentially be used as a therapeutic agent for managing S. aureus infections.
Collapse
Affiliation(s)
- Nadira Nurxat
- Department
of Laboratory Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Lili Wang
- Department
of Stomatology, Tongji Hospital, Tongji
University, Shanghai 200065, China
| | - Qichen Wang
- Department
of Laboratory Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Shujing Li
- Faculty
of Medical Laboratory Science, Shanghai
Jiao Tong University School of Medicine, 227 South Chongqing Road, Shanghai 200025, China
| | - Chen Jin
- Faculty
of Medical Laboratory Science, Shanghai
Jiao Tong University School of Medicine, 227 South Chongqing Road, Shanghai 200025, China
| | - Yaran Shi
- Faculty
of Medical Laboratory Science, Shanghai
Jiao Tong University School of Medicine, 227 South Chongqing Road, Shanghai 200025, China
| | - Ayjiamali Wulamu
- Faculty
of Medical Laboratory Science, Shanghai
Jiao Tong University School of Medicine, 227 South Chongqing Road, Shanghai 200025, China
| | - Na Zhao
- Department
of Laboratory Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yanan Wang
- Department
of Laboratory Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Hua Wang
- Department
of Laboratory Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Min Li
- Department
of Laboratory Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
- Faculty
of Medical Laboratory Science, Shanghai
Jiao Tong University School of Medicine, 227 South Chongqing Road, Shanghai 200025, China
| | - Qian Liu
- Department
of Laboratory Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| |
Collapse
|
39
|
Anderson EE, Dyzenhaus S, Ilmain JK, Sullivan MJ, van Bakel H, Torres VJ. SarS Is a Repressor of Staphylococcus aureus Bicomponent Pore-Forming Leukocidins. Infect Immun 2023; 91:e0053222. [PMID: 36939325 PMCID: PMC10112191 DOI: 10.1128/iai.00532-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/20/2023] [Indexed: 03/21/2023] Open
Abstract
Staphylococcus aureus is a successful pathogen that produces a wide range of virulence factors that it uses to subvert and suppress the immune system. These include the bicomponent pore-forming leukocidins. How the expression of these toxins is regulated is not completely understood. Here, we describe a screen to identify transcription factors involved in the regulation of leukocidins. The most prominent discovery from this screen is that SarS, a known transcription factor which had previously been described as a repressor of alpha-toxin expression, was found to be a potent repressor of leukocidins LukED and LukSF-PV. We found that inactivating sarS resulted in increased virulence both in an ex vivo model using primary human neutrophils and in an in vivo infection model in mice. Further experimentation revealed that SarS represses leukocidins by serving as an activator of Rot, a critical repressor of toxins, as well as by directly binding and repressing the leukocidin promoters. By studying contemporary clinical isolates, we identified naturally occurring mutations in the sarS promoter that resulted in overexpression of sarS and increased repression of leukocidins in USA300 bloodstream clinical isolates. Overall, these data establish SarS as an important repressor of leukocidins and expand our understanding of how these virulence factors are being regulated in vitro and in vivo by S. aureus.
Collapse
Affiliation(s)
- Exene E. Anderson
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Sophie Dyzenhaus
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Juliana K. Ilmain
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Mitchell J. Sullivan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Harm van Bakel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
40
|
Gonçalves ASC, Leitão MM, Simões M, Borges A. The action of phytochemicals in biofilm control. Nat Prod Rep 2023; 40:595-627. [PMID: 36537821 DOI: 10.1039/d2np00053a] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Covering: 2009 to 2021Antimicrobial resistance is now rising to dangerously high levels in all parts of the world, threatening the treatment of an ever-increasing range of infectious diseases. This has becoming a serious public health problem, especially due to the emergence of multidrug-resistance among clinically important bacterial species and their ability to form biofilms. In addition, current anti-infective therapies have low efficacy in the treatment of biofilm-related infections, leading to recurrence, chronicity, and increased morbidity and mortality. Therefore, it is necessary to search for innovative strategies/antibacterial agents capable of overcoming the limitations of conventional antibiotics. Natural compounds, in particular those obtained from plants, have been exhibiting promising properties in this field. Plant secondary metabolites (phytochemicals) can act as antibiofilm agents through different mechanisms of action from the available antibiotics (inhibition of quorum-sensing, motility, adhesion, and reactive oxygen species production, among others). The combination of different phytochemicals and antibiotics have revealed synergistic or additive effects in biofilm control. This review aims to bring together the most relevant reports on the antibiofilm properties of phytochemicals, as well as insights into their structure and mechanistic action against bacterial pathogens, spanning December 2008 to December 2021.
Collapse
Affiliation(s)
- Ariana S C Gonçalves
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Miguel M Leitão
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Manuel Simões
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Anabela Borges
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| |
Collapse
|
41
|
Grazul M, Balcerczak E, Sienkiewicz M. Analysis of the Presence of the Virulence and Regulation Genes from Staphylococcus aureus ( S. aureus) in Coagulase Negative Staphylococci and the Influence of the Staphylococcal Cross-Talk on Their Functions. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:5155. [PMID: 36982064 PMCID: PMC10049693 DOI: 10.3390/ijerph20065155] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/08/2023] [Accepted: 03/12/2023] [Indexed: 06/18/2023]
Abstract
Coagulase-negative staphylococci (CoNS) are increasingly becoming a public health issue worldwide due to their growing resistance to antibiotics and common involvement in complications related to invasive surgical procedures, and nosocomial and urinary tract infections. Their behavior either as a commensal or a pathogen is a result of strict regulation of colonization and virulence factors. Although functionality of virulence factors and processes involved in their regulation are quite well understood in S. aureus, little is known about them in CoNS species. Therefore, the aim of our studies was to check if clinical CoNS strains may contain virulence factors and genes involved in resistance to methicillin, that are homologous to S. aureus. Moreover, we checked the presence of elements responsible for regulation of genes that encode virulence factors typical for S. aureus in tested isolates. We also investigated whether the regulation factors produced by one CoNS isolate can affect virulence activity of other strains by co-incubation of tested isolates with supernatant from other isolates. Our studies confirmed the presence of virulence factor and regulatory genes attributed to S. aureus in CoNS isolates and indicated that one strain with an active agr gene is able to affect biofilm formation and δ-toxin activity of strains with inactive agr genes. The cognition of prevalence and regulation of virulence factors as well as antibiotic resistance of CoNS isolates is important for better control and treatment of CoNS infections.
Collapse
Affiliation(s)
- Magdalena Grazul
- Department of Pharmaceutical Microbiology and Microbiological Diagnostics, Medical University of Lodz, Muszynskiego 1 Street, 90-151 Lodz, Poland
| | - Ewa Balcerczak
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1 Street, 90-151 Lodz, Poland
| | - Monika Sienkiewicz
- Department of Pharmaceutical Microbiology and Microbiological Diagnostics, Medical University of Lodz, Muszynskiego 1 Street, 90-151 Lodz, Poland
| |
Collapse
|
42
|
Fatima M, Amin A, Alharbi M, Ishtiaq S, Sajjad W, Ahmad F, Ahmad S, Hanif F, Faheem M, Khalil AAK. Quorum Quenchers from Reynoutria japonica in the Battle against Methicillin-Resistant Staphylococcus aureus (MRSA). Molecules 2023; 28:2635. [PMID: 36985607 PMCID: PMC10056526 DOI: 10.3390/molecules28062635] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/15/2023] Open
Abstract
Over the past decade, methicillin-resistant Staphylococcus aureus (MRSA) has become a major source of biofilm formation and a major contributor to antimicrobial resistance. The genes that govern biofilm formation are regulated by a signaling mechanism called the quorum-sensing system. There is a need for new molecules to treat the infections caused by dangerous pathogens like MRSA. The current study focused on an alternative approach using juglone derivatives from Reynoutria japonica as quorum quenchers. Ten bioactive compounds from this plant, i.e., 2-methoxy-6-acetyl-7-methyljuglone, emodin, emodin 8-o-b glucoside, polydatin, resveratrol, physcion, citreorosein, quercetin, hyperoside, and coumarin were taken as ligands and docked with accessory gene regulator proteins A, B, and C and the signal transduction protein TRAP. The best ligand was selected based on docking score, ADMET properties, and the Lipinski rule. Considering all these parameters, resveratrol displayed all required drug-like properties with a docking score of -8.9 against accessory gene regulator protein C. To further assess the effectiveness of resveratrol, it was compared with the commercially available antibiotic drug penicillin. A comparison of all drug-like characteristics showed that resveratrol was superior to penicillin in many aspects. Penicillin showed a binding affinity of -6.7 while resveratrol had a score of -8.9 during docking. This was followed by molecular dynamic simulations wherein inhibitors in complexes with target proteins showed stability inside the active site during the 100 ns simulations. Structural changes due to ligand movement inside the cavity were measured in the protein targets, but they remained static due to hydrogen bonds. The results showed acceptable pharmacokinetic properties for resveratrol as compared to penicillin. Thus, we concluded that resveratrol has protective effects against Staphylococcus aureus infections and that it suppresses the quorum-sensing ability of this bacterium by targeting its infectious proteins.
Collapse
Affiliation(s)
- Maliha Fatima
- Department of Biosciences, Capital University of Science and Technology, Islamabad 44000, Pakistan
| | - Arshia Amin
- Department of Biosciences, Capital University of Science and Technology, Islamabad 44000, Pakistan
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Sundas Ishtiaq
- Department of Biosciences, Capital University of Science and Technology, Islamabad 44000, Pakistan
| | - Wasim Sajjad
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi 46000, Pakistan
| | - Faisal Ahmad
- National Center for Bioinformatics, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Sajjad Ahmad
- Department of Health and Biological Sciences, Abasyn University, Peshawar 25000, Pakistan
- Department of Computer Sciences, Virginia Tech, Blacksburg, VA 24060, USA
| | - Faisal Hanif
- Department of Microbiology Military Hospital, National University of Medical Sciences, Rawalpindi 46000, Pakistan
| | - Muhammad Faheem
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi 46000, Pakistan
| | - Atif Ali Khan Khalil
- Department of Pharmacognosy, Institute of Pharmacy, Lahore College for Women University, Lahore 54000, Pakistan
| |
Collapse
|
43
|
Kumar S, Balaya RDA, Kanekar S, Raju R, Prasad TSK, Kandasamy RK. Computational tools for exploring peptide-membrane interactions in gram-positive bacteria. Comput Struct Biotechnol J 2023; 21:1995-2008. [PMID: 36950221 PMCID: PMC10025024 DOI: 10.1016/j.csbj.2023.02.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/27/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
The vital cellular functions in Gram-positive bacteria are controlled by signaling molecules known as quorum sensing peptides (QSPs), considered promising therapeutic interventions for bacterial infections. In the bacterial system QSPs bind to membrane-coupled receptors, which then auto-phosphorylate and activate intracellular response regulators. These response regulators induce target gene expression in bacteria. One of the most reliable trends in drug discovery research for virulence-associated molecular targets is the use of peptide drugs or new functionalities. In this perspective, computational methods act as auxiliary aids for biologists, where methodologies based on machine learning and in silico analysis are developed as suitable tools for target peptide identification. Therefore, the development of quick and reliable computational resources to identify or predict these QSPs along with their receptors and inhibitors is receiving considerable attention. The databases such as Quorumpeps and Quorum Sensing of Human Gut Microbes (QSHGM) provide a detailed overview of the structures and functions of QSPs. The tools and algorithms such as QSPpred, QSPred-FL, iQSP, EnsembleQS and PEPred-Suite have been used for the generic prediction of QSPs and feature representation. The availability of compiled key resources for utilizing peptide features based on amino acid composition, positional preferences, and motifs as well as structural and physicochemical properties, including biofilm inhibitory peptides, can aid in elucidating the QSP and membrane receptor interactions in infectious Gram-positive pathogens. Herein, we present a comprehensive survey of diverse computational approaches that are suitable for detecting QSPs and QS interference molecules. This review highlights the utility of these methods for developing potential biomarkers against infectious Gram-positive pathogens.
Collapse
Key Words
- 3-HBA, 3–Hydroxybenzoic Acid
- AAC, Amino Acid Composition
- ABC, ATP-binding cassette
- ACD, Available Chemicals Database
- AIP, Autoinducing Peptide
- AMP, Anti-Microbial Peptide
- ATP, Adenosine Triphosphate
- Agr, Accessory gene regulator
- BFE, Binding Free Energy
- BIP Inhibitors
- BIP, Biofilm Inhibitory Peptides
- BLAST, Basic Local Alignment Search Tool
- BNB, Bernoulli Naïve-Bayes
- CADD, Computer-Aided Drug Design
- CSP, Competence Stimulating Peptide
- CTD, Composition-Transition-Distribution
- D, Aspartate
- DCH, 3,3′-(3,4-dichlorobenzylidene)-bis-(4-hydroxycoumarin)
- DT, Decision Tree
- FDA, Food and Drug Administration
- GBM, Gradient Boosting Machine
- GDC, g-gap Dipeptide
- GNB, Gaussian NB
- Gram-positive bacteria
- H, Histidine
- H-Kinase, Histidine Kinase
- H-phosphotransferase, Histidine Phosphotransferase
- HAM, Hamamelitannin
- HGM, Human Gut Microbiota
- HNP, Human Neutrophil Peptide
- IT, Information Theory Features
- In silico approaches
- KNN, K-Nearest Neighbors
- MCC, Mathew Co-relation Coefficient
- MD, Molecular Dynamics
- MDR, Multiple Drug Resistance
- ML, Machine Learning
- MRSA, Methicillin Resistant S. aureus
- MSL, Multiple Sequence Alignment
- OMR, Omargliptin
- OVP, Overlapping Property Features
- PCP, Physicochemical Properties
- PDB, Protein Data Bank
- PPIs, Protein-Protein Interactions
- PSM, Phenol-Soluble Modulin
- PTM, Post Translational Modification
- QS, Quorum Sensing
- QSCN, QS communication network
- QSHGM, Quorum Sensing of Human Gut Microbes
- QSI, QS Inhibitors
- QSIM, QS Interference Molecules
- QSP inhibitors
- QSP predictors
- QSP, QS Peptides
- QSPR, Quantitative Structure Property Relationship
- Quorum sensing peptides
- RAP, RNAIII-activating protein
- RF, Random Forest
- RIP, RNAIII-inhibiting peptide
- ROC, Receiver Operating Characteristic
- SAR, Structure-Activity Relationship
- SFS, Sequential Forward Search
- SIT, Sitagliptin
- SVM, Support Vector Machine
- TCS, Two-Component Sensory
- TRAP, Target of RAP
- TRG, Trelagliptin
- WHO, World Health Organization
- mRMR, minimum Redundancy and Maximum Relevance
Collapse
Affiliation(s)
- Shreya Kumar
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to be University), Mangalore 575018, India
- Centre for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India
| | | | - Saptami Kanekar
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to be University), Mangalore 575018, India
| | - Rajesh Raju
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to be University), Mangalore 575018, India
- Centre for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India
| | | | - Richard K. Kandasamy
- Centre of Molecular Inflammation Research (CEMIR), and Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Department of Laboratory Medicine and Pathology, Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
44
|
Schlievert PM, Kilgore SH, Beck LA, Yoshida T, Klingelhutz AJ, Leung DYM. Host Cationic Antimicrobial Molecules Inhibit S. aureus Exotoxin Production. mSphere 2023; 8:e0057622. [PMID: 36598227 PMCID: PMC9942567 DOI: 10.1128/msphere.00576-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 12/02/2022] [Indexed: 01/05/2023] Open
Abstract
Innate immune molecules, including antimicrobial peptides (for example, defensins) and lysozyme, function to delay or prevent bacterial infections. These molecules are commonly found on mucosal and skin surfaces. Staphylococcus aureus is a common pathogen and causes millions of infections annually. It is well known that innate immune molecules, such as defensins and lysozyme, either poorly inhibit or do not inhibit the growth of S. aureus. Our current studies show that the α-defensin human neutrophil α-defensin-1 (HNP-1) and lysozyme inhibit exotoxin production, both hemolysins and superantigens, which are required for S. aureus infection. HNP-1 inhibited exotoxin production at concentrations as low as 0.001 μg/mL. Lysozyme inhibited exotoxin production at 0.05 to 0.5 μg/mL. Both HNP-1 and lysozyme functioned through at least one two-component system (SrrA/B). The β-defensin human β-defensin 1 (HBD-1) inhibited hemolysin but not superantigen production. The cation chelator S100A8/A9 (calprotectin), compared to EDTA, was tested for the ability to inhibit exotoxin production. EDTA at high concentrations inhibited exotoxin production; these were the same concentrations that interfered with staphylococcal growth. S100A8/A9 at the highest concentration tested (10 μg/mL) had no effect on S. aureus growth but enhanced exotoxin production. Lower concentrations had no effect on growth or exotoxin production. Lysostaphin is regularly used to lyse S. aureus. The lytic concentrations of lysostaphin were the only concentrations that also inhibited growth and exotoxin production. Our studies demonstrate that a major activity of innate defensin peptides and lysozyme is inhibition of staphylococcal exotoxin production but not inhibition of growth. IMPORTANCE Staphylococcus aureus causes large numbers of both relatively benign and serious human infections, which are mediated in large part by the organisms' secreted exotoxins. Since 1921, it has been known that lysozyme and, as shown later in the 1900s, other innate immune peptides, including human neutrophil α-defensin-1 (HNP-1) and human β-defensin 1 (HBD-1), are either not antistaphylococcal or are only weakly inhibitory to growth. Our study confirms those findings but, importantly, shows that at subgrowth inhibitory concentrations, these positively charged innate immune peptides inhibit exotoxin production, including both hemolysins and the superantigen toxic shock syndrome toxin-1. The data show that the principal activity of innate immune peptides in the host is likely to be inhibition of exotoxin production required for staphylococcal mucosal or skin colonization rather than growth inhibition.
Collapse
Affiliation(s)
- Patrick M. Schlievert
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Samuel H. Kilgore
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Lisa A. Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| | - Takeshi Yoshida
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| | - Aloysius J. Klingelhutz
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | | |
Collapse
|
45
|
Dyzenhaus S, Sullivan MJ, Alburquerque B, Boff D, van de Guchte A, Chung M, Fulmer Y, Copin R, Ilmain JK, O'Keefe A, Altman DR, Stubbe FX, Podkowik M, Dupper AC, Shopsin B, van Bakel H, Torres VJ. MRSA lineage USA300 isolated from bloodstream infections exhibit altered virulence regulation. Cell Host Microbe 2023; 31:228-242.e8. [PMID: 36681080 PMCID: PMC9911362 DOI: 10.1016/j.chom.2022.12.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 10/18/2022] [Accepted: 12/02/2022] [Indexed: 01/22/2023]
Abstract
The epidemic community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) USA300 lineage has recently become a leading cause of hospital-associated bloodstream infections (BSIs). Here, we leveraged this recent introduction into hospitals and the limited genetic variation across USA300 isolates to identify mutations that contribute to its success in a new environment. We found that USA300 BSI isolates exhibit altered virulence regulation. Using comparative genomics to delineate the genes involved in this phenotype, we discovered repeated and independent mutations in the transcriptional regulator sarZ. Mutations in sarZ resulted in increased virulence of USA300 BSI isolates in a murine model of BSI. The sarZ mutations derepressed the expression and production of the surface protein ClfB, which was critical for the pathogenesis of USA300 BSI isolates. Altogether, these findings highlight ongoing evolution of a major MRSA lineage and suggest USA300 strains can optimize their fitness through altered regulation of virulence.
Collapse
Affiliation(s)
- Sophie Dyzenhaus
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Mitchell J Sullivan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bremy Alburquerque
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Daiane Boff
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Adriana van de Guchte
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Marilyn Chung
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yi Fulmer
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Richard Copin
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Juliana K Ilmain
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Anna O'Keefe
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Deena R Altman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - François-Xavier Stubbe
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Magdalena Podkowik
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Amy C Dupper
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bo Shopsin
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA; Division of Infectious Diseases and Immunology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Harm van Bakel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Victor J Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
46
|
Characterization of MroQ-Dependent Maturation and Export of the Staphylococcus aureus Accessory Gene Regulatory System Autoinducing Peptide. Infect Immun 2022; 90:e0026322. [PMID: 36073934 PMCID: PMC9584314 DOI: 10.1128/iai.00263-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Gram-positive bacteria produce small autoinducing peptides (AIPs), which act to regulate expression of genes that promote adaptive traits, including virulence. The Gram-positive pathogen Staphylococcus aureus generates a cyclic AIP that controls expression of virulence factors via the accessory gene regulatory (Agr) system. S. aureus strains belong to one of four Agr groups (Agr-I, -II, -III, and -IV); each group harbors allelic variants of AgrD, the precursor of AIP. In a prior screen for S. aureus virulence factors, we identified MroQ, a putative peptidase. A ΔmroQ mutant closely resembled a Δagr mutant and had significant defects in AIP production in an Agr-I strain. Here, we show that expression of AgrD-I in a ΔmroQ mutant leads to accumulation of an AIP processing intermediate at the membrane that coincides with a loss of secreted mature AIP, indicating that MroQ promotes maturation of AgrD-I. MroQ is conserved in all Agr sequence variants, suggesting either identical function among all Agr types or activity specific to Agr-I strains. Our data indicate that MroQ is required for AIP maturation and activity in Agr-I, -II, and -IV strains irrespective of background. However, MroQ is not required for Agr-III activity despite an identifiable role in peptide maturation. Isogenic Δagr and Δagr ΔmroQ strains complemented with Agr-I to -IV validated the critical role of MroQ in the generation of active AIP-I, -II, and -IV but not AIP-III. These findings were reinforced by skin infection studies with mice. Our data substantiate the prevailing model that MroQ is a mediator of cyclic peptide maturation.
Collapse
|
47
|
Ongpipattanakul C, Desormeaux EK, DiCaprio A, van der Donk WA, Mitchell DA, Nair SK. Mechanism of Action of Ribosomally Synthesized and Post-Translationally Modified Peptides. Chem Rev 2022; 122:14722-14814. [PMID: 36049139 PMCID: PMC9897510 DOI: 10.1021/acs.chemrev.2c00210] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ribosomally synthesized and post-translationally modified peptides (RiPPs) are a natural product class that has undergone significant expansion due to the rapid growth in genome sequencing data and recognition that they are made by biosynthetic pathways that share many characteristic features. Their mode of actions cover a wide range of biological processes and include binding to membranes, receptors, enzymes, lipids, RNA, and metals as well as use as cofactors and signaling molecules. This review covers the currently known modes of action (MOA) of RiPPs. In turn, the mechanisms by which these molecules interact with their natural targets provide a rich set of molecular paradigms that can be used for the design or evolution of new or improved activities given the relative ease of engineering RiPPs. In this review, coverage is limited to RiPPs originating from bacteria.
Collapse
Affiliation(s)
- Chayanid Ongpipattanakul
- Department of Biochemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
| | - Emily K. Desormeaux
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
| | - Adam DiCaprio
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
| | - Wilfred A. van der Donk
- Department of Biochemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
- Department of Howard Hughes Medical Institute, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
- Departments of Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, Illinois 61801, USA
| | - Douglas A. Mitchell
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
- Department of Microbiology, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
- Departments of Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, Illinois 61801, USA
| | - Satish K. Nair
- Department of Biochemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
- Departments of Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, Illinois 61801, USA
| |
Collapse
|
48
|
Wang Z, Wang Y, Wang Y, Chen W, Ji Q. CRISPR/Cpf1-Mediated Multiplex and Large-Fragment Gene Editing in Staphylococcus aureus. ACS Synth Biol 2022; 11:3049-3057. [PMID: 36001082 DOI: 10.1021/acssynbio.2c00248] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Staphylococcus aureus is a major human pathogen that causes a variety of infections, including life-threatening diseases. Research on S. aureus is constrained by complex and limited genetic manipulation methods. Here, we report a CRISPR/Cpf1-mediated system, pCpfSA, for rapid and versatile genome editing in S. aureus. In direct comparison with the existing CRISPR/Cas9-mediated genome-editing system, the pCpfSA system exhibits enhanced colony-forming units (CFUs) after editing and an expanded targetable range with comparable editing efficiency. Given the precursor crRNA (pre-crRNA) processing activity of Cpf1, the pCpfSA system also allows multiplex gene editing and large-fragment DNA knockout simply by introducing two crRNAs and the corresponding donor templates, which is difficult to achieve using the CRISPR/Cas9 system, thereby greatly expanding the genome editor toolbox for S. aureus.
Collapse
Affiliation(s)
- Zhipeng Wang
- School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Wang
- College of Bioscience and Bioengineering, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Yujue Wang
- School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Weizhong Chen
- School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Quanjiang Ji
- School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China.,Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
49
|
Rosenbergová Z, Oftedal TF, Ovchinnikov KV, Thiyagarajah T, Rebroš M, Diep DB. Identification of a Novel Two-Peptide Lantibiotic from Vagococcus fluvialis. Microbiol Spectr 2022; 10:e0095422. [PMID: 35730941 PMCID: PMC9431498 DOI: 10.1128/spectrum.00954-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 05/24/2022] [Indexed: 11/20/2022] Open
Abstract
Infections caused by multiresistant pathogens have become a major problem in both human and veterinary medicine. Due to the declining efficacy of many antibiotics, new antimicrobials are needed. Promising alternatives or additions to antibiotics are bacteriocins, antimicrobial peptides of bacterial origin with activity against many pathogens, including antibiotic-resistant strains. From a sample of fermented maize, we isolated a Vagococcus fluvialis strain producing a bacteriocin with antimicrobial activity against multiresistant Enterococcus faecium. Whole-genome sequencing revealed the genes for a novel two-peptide lantibiotic. The production of the lantibiotic by the isolate was confirmed by matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectrometry, which revealed distinct peaks at 4,009.4 m/z and 3,181.7 m/z in separate fractions from reversed-phase chromatography. The combination of the two peptides resulted in a 1,200-fold increase in potency, confirming the two-peptide nature of the bacteriocin, named vagococcin T. The bacteriocin was demonstrated to kill sensitive cells by the formation of pores in the cell membrane, and its inhibition spectrum covers most Gram-positive bacteria, including multiresistant pathogens. To our knowledge, this is the first bacteriocin characterized from Vagococcus. IMPORTANCE Enterococci are common commensals in the intestines of humans and animals, but in recent years, they have been identified as one of the major causes of hospital-acquired infections due to their ability to quickly acquire virulence and antibiotic resistance determinants. Many hospital isolates are multiresistant, thereby making current therapeutic options critically limited. Novel antimicrobials or alternative therapeutic approaches are needed to overcome this global problem. Bacteriocins, natural ribosomally synthesized peptides produced by bacteria to eliminate other bacterial species living in a competitive environment, provide such an alternative. In this work, we purified and characterized a novel two-peptide lantibiotic produced by Vagococcus fluvialis LMGT 4216 isolated from fermented maize. The novel lantibiotic showed a broad spectrum of inhibition of Gram-positive strains, including vancomycin-resistant Enterococcus faecium, demonstrating its therapeutic potential.
Collapse
Affiliation(s)
- Zuzana Rosenbergová
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
- Institute of Biotechnology, Faculty of Chemical and Food Technology, Slovak University of Technology, Bratislava, Slovakia
| | - Thomas F. Oftedal
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Kirill V. Ovchinnikov
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Thasanth Thiyagarajah
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Martin Rebroš
- Institute of Biotechnology, Faculty of Chemical and Food Technology, Slovak University of Technology, Bratislava, Slovakia
| | - Dzung B. Diep
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| |
Collapse
|
50
|
Sahreen S, Mukhtar H, Imre K, Morar A, Herman V, Sharif S. Exploring the Function of Quorum Sensing Regulated Biofilms in Biological Wastewater Treatment: A Review. Int J Mol Sci 2022; 23:ijms23179751. [PMID: 36077148 PMCID: PMC9456111 DOI: 10.3390/ijms23179751] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/21/2022] [Accepted: 08/26/2022] [Indexed: 11/26/2022] Open
Abstract
Quorum sensing (QS), a type of bacterial cell–cell communication, produces autoinducers which help in biofilm formation in response to cell population density. In this review, biofilm formation, the role of QS in biofilm formation and development with reference to biological wastewater treatment are discussed. Autoinducers, for example, acyl-homoserine lactones (AHLs), auto-inducing oligo-peptides (AIPs) and autoinducer 2, present in both Gram-negative and Gram-positive bacteria, with their mechanism, are also explained. Over the years, wastewater treatment (WWT) by QS-regulated biofilms and their optimization for WWT have gained much attention. This article gives a comprehensive review of QS regulation methods, QS enrichment methods and QS inhibition methods in biological waste treatment systems. Typical QS enrichment methods comprise adding QS molecules, adding QS accelerants and cultivating QS bacteria, while typical QS inhibition methods consist of additions of quorum quenching (QQ) bacteria, QS-degrading enzymes, QS-degrading oxidants, and QS inhibitors. Potential applications of QS regulated biofilms for WWT have also been summarized. At last, the knowledge gaps present in current researches are analyzed, and future study requirements are proposed.
Collapse
Affiliation(s)
- Sania Sahreen
- Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan
| | - Hamid Mukhtar
- Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan
- Correspondence: (H.M.); (K.I.); Tel.: +92-3334245581 (H.M.); +40-256277186 (K.I.)
| | - Kálmán Imre
- Department of Animal Production and Veterinary Public Health, Faculty of Veterinary Medicine, Banat’s University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania”, 300645 Timisoara, Romania
- Correspondence: (H.M.); (K.I.); Tel.: +92-3334245581 (H.M.); +40-256277186 (K.I.)
| | - Adriana Morar
- Department of Animal Production and Veterinary Public Health, Faculty of Veterinary Medicine, Banat’s University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania”, 300645 Timisoara, Romania
| | - Viorel Herman
- Department of Infectious Diseases and Preventive Medicine, Faculty of Veterinary Medicine, Banat’s University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania”, 300645 Timisoara, Romania
| | - Sundas Sharif
- Institute of Industrial Biotechnology, Government College University, Lahore 54000, Pakistan
| |
Collapse
|