1
|
Flores C, Millard S, Seekatz AM. Bridging Ecology and Microbiomes: Applying Ecological Theories in Host-associated Microbial Ecosystems. CURRENT CLINICAL MICROBIOLOGY REPORTS 2025; 12:9. [PMID: 40248762 PMCID: PMC12000275 DOI: 10.1007/s40588-025-00246-z] [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] [Accepted: 03/28/2025] [Indexed: 04/19/2025]
Abstract
Purpose of Review This review explores the application of classical ecological theory to host-associated microbiomes during initial colonization, maintenance, and recovery. We discuss unique challenges of applying these theories to host-associated microbiomes and host factors to consider going forward. Recent Findings Recent studies applying community ecology principles to host microbiomes continue to demonstrate a role for both selective and stochastic processes in shaping host-associated microbiomes. However, ecological frameworks developed to describe dynamics during homeostasis do not necessarily apply during diseased or highly perturbed states, where large variations can potentially lead to alternate stable states. Summary Despite providing valuable insights, the application of ecological theories to host-associated microbiomes has some unique challenges. The integration of host-specific factors, such as genotype or immune dynamics in ecological models or frameworks is crucial for understanding host microbiome assembly and stability, which could improve our ability to predict microbiome outcomes and improve host health.
Collapse
Affiliation(s)
- Clara Flores
- Department of Biological Sciences, Clemson University, Life Sciences Building 157 A, 190 Collings St, Clemson, SC 29634 USA
| | - Sophie Millard
- Department of Biological Sciences, Clemson University, Life Sciences Building 157 A, 190 Collings St, Clemson, SC 29634 USA
| | - Anna M. Seekatz
- Department of Biological Sciences, Clemson University, Life Sciences Building 157 A, 190 Collings St, Clemson, SC 29634 USA
| |
Collapse
|
2
|
Kals M, Kals E, Kotar J, Donald A, Mancini L, Cicuta P. Antibiotics change the population growth rate heterogeneity and morphology of bacteria. PLoS Pathog 2025; 21:e1012924. [PMID: 39908318 PMCID: PMC11835381 DOI: 10.1371/journal.ppat.1012924] [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: 06/21/2024] [Revised: 02/18/2025] [Accepted: 01/23/2025] [Indexed: 02/07/2025] Open
Abstract
A better understanding of the system-level effects of antibiotics on bacterial cells is essential to address the growing challenge of antibiotic resistance. Utilising Multipad Agarose Plate (MAP) platforms, we monitor the growth rate and cell morphology of three clinically relevant species (E.coli, S.aureus and P.aeruginosa) following exposure to 14 antibiotics across 11 concentrations (31 microbe-antibiotic combinations in total). Our results reveal a consistent increase in population growth rate heterogeneity (PGRH) as drug concentrations approach the minimum inhibitory concentration (MIC). Strikingly, the magnitude of this heterogeneity correlates with the functional distance between the ribosome and the specific cellular processes targeted by the antibiotics. Among the seven antibiotic classes studied, protein synthesis inhibitors and disruptors cause the lowest PGRH, while heterogeneity progressively increases with RNA synthesis inhibitors, DNA replication inhibitors, cell membrane disruptors and cell wall synthesis inhibitors. Because the ribosome is central to growth rate control, we hypothesize that heterogeneity might arise at the system level as a result of the propagation of damage to protein synthesis. Low heterogeneity is desirable from a clinical perspective, as high heterogeneity is often associated with persistence and treatment survival. Additionally, we observed a strong correlation between morphological alterations and growth inhibition across all antibiotics and species tested. This led to the development of a novel morphological parameter, MOR50, which enables rapid estimation of MIC for antibiotic susceptibility testing (AST) with a single snapshot after just 2.5 hours of incubation. In addition to introducing a novel, resource-efficient and rapid AST method, our findings shed new light on the system-level effects of antibiotic perturbations on bacteria, which might inform treatment design.
Collapse
Affiliation(s)
- Morten Kals
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
- Synoptics Ltd., Cambridge, United Kingdom
| | - Emma Kals
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Jurij Kotar
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| | | | - Leonardo Mancini
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Pietro Cicuta
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
3
|
Felicianna, Lo EKK, Chen C, Ismaiah MJ, Zhang F, Leung HKM, El-Nezami H. Low-dose valine attenuates diet-induced metabolic dysfunction-associated steatotic liver disease (MASLD) in mice by enhancing leptin sensitivity and modulating the gut microbiome. Mol Metab 2024; 90:102059. [PMID: 39489290 PMCID: PMC11616088 DOI: 10.1016/j.molmet.2024.102059] [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: 08/21/2024] [Revised: 10/10/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024] Open
Abstract
OBJECTIVES Elevated circulating branched-chain amino acids (BCAAs) have been associated with obesity, insulin resistance, and MASLD. Nonetheless, BCAA supplementation has been shown to provide protective outcomes towards the intervention of MASLD. Currently, there is a lack of study towards the contribution of the BCAA: valine on MASLD. Herein, the effect of low-dose valine supplementation was investigated for its role in the progression of MASLD. METHODS C57BL/6J mice were fed a high-fat/high-cholesterol diet (HFD) to induce MASLD. Upon the establishment of MASLD, valine was supplemented via voluntary oral administration. Clinical and biochemical parameters associated with MASLD were measured, and molecular mechanism and gut microbiota modulation from the effect of valine were investigated. RESULTS Low-dose valine was found to attenuate the progression of MASLD, significantly reducing the gain in body weight, liver weight, and epididymal white adipose tissue (eWAT) weight, while also attenuating hyperglycemia and hyperleptinemia, and improving serum lipid profiles. Mechanistically, in the liver, genes related to hepatic lipogenesis and cholesterol biosynthesis were downregulated, while those associated with fatty acid oxidation, autophagy, and antioxidant capacity were upregulated, and AMPK pathway activity was enhanced. Liver and hypothalamic leptin resistance and inflammation were also attenuated, allowing better appetite control in mice fed a HFD and leading to reduced food intake. Additionally, metabolic flexibility in the eWAT was improved, and the gut microbiome was modulated by low-dose valine supplementation. CONCLUSION Low-dose valine supplementation attenuates MASLD by enhancing systemic leptin sensitivity and modulating the gut microbiome.
Collapse
Affiliation(s)
- Felicianna
- School of Biological Sciences, University of Hong Kong, Pokfulam 999077, Hong Kong, China
| | - Emily K K Lo
- School of Biological Sciences, University of Hong Kong, Pokfulam 999077, Hong Kong, China
| | - Congjia Chen
- School of Biological Sciences, University of Hong Kong, Pokfulam 999077, Hong Kong, China
| | - Marsena J Ismaiah
- School of Biological Sciences, University of Hong Kong, Pokfulam 999077, Hong Kong, China
| | - Fangfei Zhang
- School of Biological Sciences, University of Hong Kong, Pokfulam 999077, Hong Kong, China
| | - Hoi Kit Matthew Leung
- School of Biological Sciences, University of Hong Kong, Pokfulam 999077, Hong Kong, China
| | - Hani El-Nezami
- School of Biological Sciences, University of Hong Kong, Pokfulam 999077, Hong Kong, China; Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland.
| |
Collapse
|
4
|
Ceylan A, Onal I, Aydin FO, Mergen B, Yildirim Y. Improvement of Clinical Findings, Meibography and Tear Film Parameters in Pediatric Ocular Rosacea Patients After a Standard Treatment Protocol. Ocul Immunol Inflamm 2024; 32:2130-2137. [PMID: 38512290 DOI: 10.1080/09273948.2024.2328791] [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: 10/06/2023] [Revised: 02/26/2024] [Accepted: 03/05/2024] [Indexed: 03/22/2024]
Abstract
PURPOSE The objective of this study was to illustrate the changes in ocular findings, meibography, and tear break-up time (TBUT) values in pediatric patients with ocular rosacea following a standardized treatment. METHODS The study included consecutive patients diagnosed with ocular rosacea, referred to a tertiary hospital between 2021 and 2023. Each patient underwent biomicroscopic examinations, non-invasive TBUT assessments, corneal fluorescein staining (evaluated using the Oxford scoring system), and meibography. The standard treatment protocol involved warm compresses, eyelid hygiene, preservative-free sodium hyaluronate eye drops (administered four times daily), topical azithromycin 1.5% (twice daily for 3 days), topical steroids (loteprednol 0.5%, four times daily for 2 weeks), and either doxycycline 100 mg/day for 14 days or oral suspension of azithromycin 10 mg/kg for 3 days followed by an additional three-day course of treatment administered 10 days later (for patients above and below 14 years of age, respectively). RESULTS The study included 18 patients, with 10 (55.5%) being female and 8 (44.4%) being male, with a mean age of 9.7 ± 4.5 years (range: 3-18). Four patients displayed cutaneous involvement. The treatments resulted in significant improvements in the Oxford scores, reduction in corneal neovascularization, and increased TBUT (p < 0.001, p = 0.016, p < 0.001, respectively). Meibomian gland loss area also significantly improved post-treatment (27.4 ± 6.7% vs 39.2 ± 13.4%, p = 0.001). CONCLUSION This study demonstrated that pediatric ocular rosacea patients may exhibit improved meibomian gland function, regression of corneal neovascularization, and enhanced tear film parameters following a standardized treatment protocol that includes both topical and systemic approaches.
Collapse
Affiliation(s)
- Ali Ceylan
- Department of Ophthalmology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Irem Onal
- Department of Ophthalmology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Fahri Onur Aydin
- Department of Ophthalmology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Burak Mergen
- Department of Ophthalmology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Yusuf Yildirim
- Department of Ophthalmology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| |
Collapse
|
5
|
Tewari N, Dey P. Navigating commensal dysbiosis: Gastrointestinal host-pathogen interplay orchestrating opportunistic infections. Microbiol Res 2024; 286:127832. [PMID: 39013300 DOI: 10.1016/j.micres.2024.127832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/23/2024] [Accepted: 07/01/2024] [Indexed: 07/18/2024]
Abstract
The gut commensals, which are usually symbiotic or non-harmful bacteria that live in the gastrointestinal tract, have a positive impact on the health of the host. This review, however, specifically discuss distinct conditions where commensals aid in the development of pathogenic opportunistic infections. We discuss that the categorization of gut bacteria as either pathogens or non-pathogens depends on certain circumstances, which are significantly affected by the tissue microenvironment and the dynamic host-microbe interaction. Under favorable circumstances, commensals have the ability to transform into opportunistic pathobionts by undergoing overgrowth. These conditions include changes in the host's physiology, simultaneous infection with other pathogens, effective utilization of nutrients, interactions between different species of bacteria, the formation of protective biofilms, genetic mutations that enhance pathogenicity, acquisition of genes associated with virulence, and the ability to avoid the host's immune response. These processes allow commensals to both initiate infections themselves and aid other pathogens in populating the host. This review highlights the need of having a detailed and sophisticated knowledge of the two-sided nature of gut commensals. Although commensals mostly promote health, they may also become harmful in certain changes in the environment or the body's functioning. This highlights the need of acknowledging the intricate equilibrium in interactions between hosts and microbes, which is crucial for preserving intestinal homeostasis and averting diseases. Finally, we also emphasize the further need of research to better understand and anticipate the behavior of gut commensals in different situations, since they play a crucial and varied role in human health and disease.
Collapse
Affiliation(s)
- Nisha Tewari
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab 147004, India
| | - Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab 147004, India.
| |
Collapse
|
6
|
Williams A, Sinanaj B, Hoysted GA. Plant-microbe interactions through a lens: tales from the mycorrhizosphere. ANNALS OF BOTANY 2024; 133:399-412. [PMID: 38085925 PMCID: PMC11006548 DOI: 10.1093/aob/mcad191] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 12/11/2023] [Indexed: 04/12/2024]
Abstract
BACKGROUND The soil microbiome plays a pivotal role in maintaining ecological balance, supporting food production, preserving water quality and safeguarding human health. Understanding the intricate dynamics within the soil microbiome necessitates unravelling complex bacterial-fungal interactions (BFIs). BFIs occur in diverse habitats, such as the phyllosphere, rhizosphere and bulk soil, where they exert substantial influence on plant-microbe associations, nutrient cycling and overall ecosystem functions. In various symbiotic associations, fungi form mycorrhizal connections with plant roots, enhancing nutrient uptake through the root and mycorrhizal pathways. Concurrently, specific soil bacteria, including mycorrhiza helper bacteria, play a pivotal role in nutrient acquisition and promoting plant growth. Chemical communication and biofilm formation further shape plant-microbial interactions, affecting plant growth, disease resistance and nutrient acquisition processes. SCOPE Promoting synergistic interactions between mycorrhizal fungi and soil microbes holds immense potential for advancing ecological knowledge and conservation. However, despite the significant progress, gaps remain in our understanding of the evolutionary significance, perception, functional traits and ecological relevance of BFIs. Here we review recent findings obtained with respect to complex microbial communities - particularly in the mycorrhizosphere - and include the latest advances in the field, outlining their profound impacts on our understanding of ecosystem dynamics and plant physiology and function. CONCLUSIONS Deepening our understanding of plant BFIs can help assess their capabilities with regard to ecological and agricultural safe-guarding, in particular buffering soil stresses, and ensuring sustainable land management practices. Preserving and enhancing soil biodiversity emerge as critical imperatives in sustaining life on Earth amidst pressures of anthropogenic climate change. A holistic approach integrates scientific knowledge on bacteria and fungi, which includes their potential to foster resilient soil ecosystems for present and future generations.
Collapse
Affiliation(s)
- Alex Williams
- Plants, Photosynthesis and Soil, School of Bioscience, University of Sheffield, Sheffield, S10 2TN, UK
| | - Besiana Sinanaj
- Plants, Photosynthesis and Soil, School of Bioscience, University of Sheffield, Sheffield, S10 2TN, UK
| | - Grace A Hoysted
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland
| |
Collapse
|
7
|
Bertrand BP, Shinde D, Thomas VC, Whiteley M, Ibberson CB, Kielian T. Metabolic diversity of human macrophages: potential influence on Staphylococcus aureus intracellular survival. Infect Immun 2024; 92:e0047423. [PMID: 38179975 PMCID: PMC10863412 DOI: 10.1128/iai.00474-23] [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: 11/14/2023] [Accepted: 12/04/2023] [Indexed: 01/06/2024] Open
Abstract
Staphylococcus aureus is a leading cause of medical device-associated biofilm infections. This is influenced by the ability of S. aureus biofilm to evade the host immune response, which is partially driven by the anti-inflammatory cytokine interleukin-10 (IL-10). Here, we show that treatment of human monocyte-derived macrophages (HMDMs) with IL-10 enhanced biofilm formation, suggesting that macrophage anti-inflammatory programming likely plays an important role during the transition from planktonic to biofilm growth. To identify S. aureus genes that were important for intracellular survival in HMDMs and how this was affected by IL-10, transposon sequencing was performed. The size of the S. aureus essential genome was similar between unstimulated HMDMs and the outgrowth control (18.5% vs 18.4%, respectively, with 54.4% overlap) but increased to 22.5% in IL-10-treated macrophages, suggesting that macrophage polarization status exerts differential pressure on S. aureus. Essential genes for S. aureus survival within IL-10-polarized HMDMs were dominated by negative regulatory pathways, including nitrogen and RNA metabolism, whereas S. aureus essential genes within untreated HMDMs were enriched in biosynthetic pathways such as purine and pyrimidine biosynthesis. To explore how IL-10 altered the macrophage intracellular metabolome, targeted metabolomics was performed on HMDMs from six individual donors. IL-10 treatment led to conserved alterations in distinct metabolites that were increased (dihydroxyacetone phosphate, glyceraldehyde-3-phosphate, and acetyl-CoA) or reduced (fructose-6-phosphate, aspartic acid, and ornithine) across donors, whereas other metabolites were variable. Collectively, these findings highlight an important aspect of population-level heterogeneity in human macrophage responsiveness that should be considered when translating results to a patient population.IMPORTANCEOne mechanism that Staphylococcus aureus biofilm elicits in the host to facilitate infection persistence is the production of the anti-inflammatory cytokine interleukin-10 (IL-10). Here, we show that exposure of human monocyte-derived macrophages (HMDMs) to IL-10 promotes S. aureus biofilm formation and programs intracellular bacteria to favor catabolic pathways. Examination of intracellular metabolites in HMDMs revealed heterogeneity between donors that may explain the observed variability in essential genes for S. aureus survival based on nutrient availability for bacteria within the intracellular compartment. Collectively, these studies provide novel insights into how IL-10 polarization affects S. aureus intracellular survival in HMDMs and the importance of considering macrophage heterogeneity between human donors as a variable when examining effector mechanisms.
Collapse
Affiliation(s)
- Blake P. Bertrand
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Dhananjay Shinde
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Vinai C. Thomas
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Marvin Whiteley
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Carolyn B. Ibberson
- School of Biological Sciences, University of Oklahoma, Norman, Oklahoma, USA
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
8
|
Arbez B, Retourney C, Quilès F, Francius G, Fierobe HP, El-Kirat-Chatel S. Simultaneous enzyme grafting on bio-inspired scaffolds for antibacterial protection. MATERIALS ADVANCES 2024; 5:1171-1184. [DOI: 10.1039/d3ma00703k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Surface bacterial contamination represents a crucial health and industrial concern which requires new strategies to be continuously developed.
Collapse
Affiliation(s)
- Baptiste Arbez
- Université de Lorraine, CNRS, LCPME, 405 Rue de Vandoeuvre, 54600 Villers-lès-Nancy, France
| | - Chloé Retourney
- Université de Lorraine, CNRS, LCPME, 405 Rue de Vandoeuvre, 54600 Villers-lès-Nancy, France
| | - Fabienne Quilès
- Université de Lorraine, CNRS, LCPME, 405 Rue de Vandoeuvre, 54600 Villers-lès-Nancy, France
| | - Gregory Francius
- Université de Lorraine, CNRS, LCPME, 405 Rue de Vandoeuvre, 54600 Villers-lès-Nancy, France
| | - Henri-Pierre Fierobe
- Laboratoire de Chimie Bactérienne (LCB), CNRS, Université d'Aix-Marseille, UMR7283 31 Chemin Joseph Aiguier, 13009 Marseille, France
| | - Sofiane El-Kirat-Chatel
- Université de Lorraine, CNRS, LCPME, 405 Rue de Vandoeuvre, 54600 Villers-lès-Nancy, France
- CBMN, CNRS UMR 5248, IPB, Université de Bordeaux, Pessac, France
| |
Collapse
|
9
|
Ribeiro IS, Muniz IPR, Galantini MPL, Gonçalves CV, Lima PHB, Silva ES, Silva NR, Rosa FCS, Rosa LP, Costa DJ, Amaral JG, da Silva RAA. Characterization of Brazilian green propolis as a photosensitizer for LED light-induced antimicrobial photodynamic therapy (aPDT) against methicillin-resistant Staphylococcus aureus (MRSA) and Vancomycin-intermediate Staphylococcus aureus (VISA). Photochem Photobiol Sci 2023; 22:2877-2890. [PMID: 37923909 DOI: 10.1007/s43630-023-00495-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 10/11/2023] [Indexed: 11/06/2023]
Abstract
Staphylococcus aureus is the primary cause of skin and soft tissue infections. Its significant adaptability and the development of resistance are the main factors linked to its spread and the challenges in its treatment. Antimicrobial photodynamic therapy emerges as a promising alternative. This work aimed to characterize the antimicrobial photodynamic activity of Brazilian green propolis, along with the key bioactive compounds associated with this activity. Initially, a scanning spectrometry was conducted to assess the wavelengths with the potential to activate green propolis. Subsequently, reference strains of methicillin-resistant Staphylococcus aureus (MRSA ATCC 43300) and vancomycin-intermediate Staphylococcus aureus (VISA ATCC 700699) were exposed to varying concentrations of green propolis: 1 µg/mL, 5 µg/mL, 10 µg/mL, 50 µg /mL and 100 µg/mL and were stimulated by blue, green or red LED light. Finally, high-performance liquid chromatography coupled with a diode array detector and tandem mass spectrometry techniques, along with classic molecular networking analysis, was performed to identify potential bioactive molecules with photodynamic activity. Brazilian green propolis exhibits a pronounced absorption peak and heightened photo-responsiveness when exposed to blue light within the range of 400 nm and 450 nm. This characteristic reveals noteworthy significant photodynamic activity against MRSA and VISA at concentrations from 5 µg/mL. Furthermore, the propolis comprises compounds like curcumin and other flavonoids sourced from flavone, which possess the potential for photodynamic activity and other antimicrobial functions. Consequently, Brazilian green propolis holds promise as an excellent bactericidal agent, displaying a synergistic antibacterial property enhanced by light-induced photodynamic effects.
Collapse
Affiliation(s)
- Israel Souza Ribeiro
- Universidade Federal da Bahia, Campus Anísio Teixeira-Instituto Multidisciplinar Em Saúde, Rua Hormindo Barros, 58, Bairro Candeias, CEP: 45.029-094, Vitória da Conquista, Bahia, Brasil
- Universidade Federal Do Sul da Bahia, Campus Paulo Freire, 250 Praça Joana Angélica, Bairro São José, 45.988-058, Teixeira de Freitas, Bahia, Brasil
| | - Igor Pereira Ribeiro Muniz
- Universidade Federal da Bahia, Campus Anísio Teixeira-Instituto Multidisciplinar Em Saúde, Rua Hormindo Barros, 58, Bairro Candeias, CEP: 45.029-094, Vitória da Conquista, Bahia, Brasil
| | - Maria Poliana Leite Galantini
- Universidade Federal da Bahia, Campus Anísio Teixeira-Instituto Multidisciplinar Em Saúde, Rua Hormindo Barros, 58, Bairro Candeias, CEP: 45.029-094, Vitória da Conquista, Bahia, Brasil
| | - Caroline Vieira Gonçalves
- Universidade Federal da Bahia, Campus Anísio Teixeira-Instituto Multidisciplinar Em Saúde, Rua Hormindo Barros, 58, Bairro Candeias, CEP: 45.029-094, Vitória da Conquista, Bahia, Brasil
| | - Paulo Henrique Bispo Lima
- Universidade Federal da Bahia, Campus Anísio Teixeira-Instituto Multidisciplinar Em Saúde, Rua Hormindo Barros, 58, Bairro Candeias, CEP: 45.029-094, Vitória da Conquista, Bahia, Brasil
| | - Emely Soares Silva
- Universidade Federal da Bahia, Campus Anísio Teixeira-Instituto Multidisciplinar Em Saúde, Rua Hormindo Barros, 58, Bairro Candeias, CEP: 45.029-094, Vitória da Conquista, Bahia, Brasil
| | - Nathalia Rosa Silva
- Universidade Federal da Bahia, Campus Anísio Teixeira-Instituto Multidisciplinar Em Saúde, Rua Hormindo Barros, 58, Bairro Candeias, CEP: 45.029-094, Vitória da Conquista, Bahia, Brasil
| | - Francine Cristina Silva Rosa
- Universidade Federal da Bahia, Campus Anísio Teixeira-Instituto Multidisciplinar Em Saúde, Rua Hormindo Barros, 58, Bairro Candeias, CEP: 45.029-094, Vitória da Conquista, Bahia, Brasil
| | - Luciano Pereira Rosa
- Universidade Federal da Bahia, Campus Anísio Teixeira-Instituto Multidisciplinar Em Saúde, Rua Hormindo Barros, 58, Bairro Candeias, CEP: 45.029-094, Vitória da Conquista, Bahia, Brasil
| | - Dirceu Joaquim Costa
- Universidade Estadual Do Sudoeste da Bahia, Campus Vitória da Conquista, Av. Edmundo Silveira Flores, 27-43-Lot, Alto da Boa Vista, CEP: 45029-066, Vitória da Conquista, Bahia, Brasil
| | - Juliano Geraldo Amaral
- Universidade Federal da Bahia, Campus Anísio Teixeira-Instituto Multidisciplinar Em Saúde, Rua Hormindo Barros, 58, Bairro Candeias, CEP: 45.029-094, Vitória da Conquista, Bahia, Brasil
| | - Robson Amaro Augusto da Silva
- Universidade Federal da Bahia, Campus Anísio Teixeira-Instituto Multidisciplinar Em Saúde, Rua Hormindo Barros, 58, Bairro Candeias, CEP: 45.029-094, Vitória da Conquista, Bahia, Brasil.
| |
Collapse
|
10
|
Dey P, Ray Chaudhuri S. The opportunistic nature of gut commensal microbiota. Crit Rev Microbiol 2023; 49:739-763. [PMID: 36256871 DOI: 10.1080/1040841x.2022.2133987] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 07/30/2022] [Accepted: 10/05/2022] [Indexed: 11/03/2022]
Abstract
The abundance of gut commensals has historically been associated with health-promoting effects despite the fact that the definition of good or bad microbiota remains condition-specific. The beneficial or pathogenic nature of microbiota is generally dictated by the dimensions of host-microbiota and microbe-microbe interactions. With the increasing popularity of gut microbiota in human health and disease, emerging evidence suggests opportunistic infections promoted by those gut bacteria that are generally considered beneficial. Therefore, the current review deals with the opportunistic nature of the gut commensals and aims to summarise the concepts behind the occasional commensal-to-pathogenic transformation of the gut microbes. Specifically, relevant clinical and experimental studies have been discussed on the overgrowth and bacteraemia caused by commensals. Three key processes and their underlying mechanisms have been summarised to be responsible for the opportunistic nature of commensals, viz. improved colonisation fitness that is dictated by commensal-pathogen interactions and availability of preferred nutrients; pathoadaptive mutations that can trigger the commensal-to-pathogen transformation; and evasion of host immune response as a survival and proliferation strategy of the microbes. Collectively, this review provides an updated concept summary on the underlying mechanisms of disease causative events driven by gut commensal bacteria.
Collapse
Affiliation(s)
- Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Saumya Ray Chaudhuri
- Council of Scientific and Industrial Research (CSIR), Institute of Microbial Technology, Chandigarh, India
| |
Collapse
|
11
|
Arya R, Kim T, Youn JW, Bae T, Kim KK. Identification of an antivirulence agent targeting the master regulator of virulence genes in Staphylococcus aureus. Front Cell Infect Microbiol 2023; 13:1268044. [PMID: 38029271 PMCID: PMC10644738 DOI: 10.3389/fcimb.2023.1268044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
The emergence of bactericidal antibiotic-resistant strains has increased the demand for alternative therapeutic agents, such as antivirulence agents targeting the virulence regulators of pathogens. Staphylococcus aureus exoprotein expression (sae) locus, the master regulator of virulence gene expression in multiple drug-resistant S. aureus, is a promising therapeutic target. In this study, we screened a small-molecule library using a SaeRS green fluorescent protein (GFP)-reporter that responded to transcription controlled by the sae locus. We identified the compound, N-(2-methylcyclohexyl)-11-oxo-10,11-dihydrodibenzo[b,f][1,4]thiazepine-8-carboxamide (SKKUCS), as an efficient repressor of sae-regulated GFP activity. SKKUCS inhibited hemolysin production and reduced α-hemolysin-mediated cell lysis. Moreover, SKKUCS substantially reduced the expression levels of various virulence genes controlled by the master regulators, sae, and the accessory gene regulator (agr), demonstrating its potential as an antivirulence reagent targeting the key virulence regulators. Furthermore, autokinase inhibition assay and molecular docking suggest that SKKUCS inhibits the kinase activity of SaeS and potentially targets the active site of SaeS kinase, possibly inhibiting ATP binding. Next, we evaluated the efficacy and toxicity of SKKUCS in vivo using murine models of staphylococcal intraperitoneal and skin infections. Treatment with SKKUCS markedly increased animal survival and significantly decreased the bacterial burden in organs and skin lesion sizes. These findings highlight SKKUCS as a potential antivirulence drug for drug-resistant staphylococcal infections.
Collapse
Affiliation(s)
- Rekha Arya
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Institute of Antibacterial Resistance Research and Therapeutics, Suwon, Republic of Korea
- Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Truc Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Institute of Antibacterial Resistance Research and Therapeutics, Suwon, Republic of Korea
| | - Joo Won Youn
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Institute of Antibacterial Resistance Research and Therapeutics, Suwon, Republic of Korea
| | - Taeok Bae
- Department of Microbiology and Immunology, Indiana University School of Medicine-Northwest, Gary, IN, United States
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Institute of Antibacterial Resistance Research and Therapeutics, Suwon, Republic of Korea
| |
Collapse
|
12
|
Du J, Cong Y, Wang X, Kang Y, Zhang P, Li L. Green Synthesis of Antimicrobial Peptide-Protected Silver Nanoclusters with Regulated Antibacterial Behavior. ACS APPLIED BIO MATERIALS 2023; 6:3919-3926. [PMID: 37678239 DOI: 10.1021/acsabm.3c00646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
The antibacterial system based on the silver element has been a very promising antibacterial material. However, the antibacterial activity of silver nanomaterials largely depends on their chemical composition and physical properties. Herein, we prepared ultrasmall silver nanoclusters by directly reducing silver ions with antimicrobial peptide in a green way. The positively charged peptide ligands drove the silver nanoclusters binding to bacteria by electrostatic attraction. Contrary to the large-sized silver nanomaterials, the ultrasmall silver nanoclusters were able to rapidly penetrate bacteria membranes via strong hydrophobic association, significantly promoting the generation of reactive oxygen species, and the subsequent high oxidative stress led to bacterial death. Moreover, the silver nanoclusters with antibacterial peptide ligand exhibited good stability, low cytotoxicity, and long-term antibacterial activity. Besides, synergistic enhancement of silver nanoclusters on antibacterial activity was observed. Therefore, the silver nanoclusters conjugated with the antimicrobial peptide can act as a synergistic antibacterial agent, in which their bio-interactions with bacteria have been regulated to achieve a rapid, long-lasting, and broad-spectrum antibacterial effect.
Collapse
Affiliation(s)
- Jingrui Du
- State Key Laboratory for Advanced Metals and Materials, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, P. R. China
| | - Yujie Cong
- State Key Laboratory for Advanced Metals and Materials, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, P. R. China
| | - Xiaoyu Wang
- State Key Laboratory for Advanced Metals and Materials, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, P. R. China
| | - Yuetong Kang
- State Key Laboratory for Advanced Metals and Materials, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, P. R. China
| | - Pengbo Zhang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, P. R. China
| | - Lidong Li
- State Key Laboratory for Advanced Metals and Materials, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, P. R. China
| |
Collapse
|
13
|
Bayan R, Tauseef I, Hussain M, Ahmed MS, Haider A, Khalil AA, Islam SU, Subhan F. Fish collagen peptides' modulating effect on human skin microbiota against pathogenic Staphylococcus aureus. Future Microbiol 2023; 18:795-807. [PMID: 37650688 DOI: 10.2217/fmb-2022-0208] [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: 09/01/2023] Open
Abstract
Aim: The current research aims to design effective strategies to enhance the body's immune system against pathogenic bacteria. Methods: Skin commensals were isolated, identified and cultured in fish collagen peptides (FCPs). Results: After culturing in FCP, the skin commensals were used in a dose-dependent manner for Staphylococcus aureus in a dual-culture test, which showed significant growth inhibition of the pathogenic bacteria, which concluded that FCP induced the immune defense system of skin microbiota against pathogenic strains. Conclusion: Results have validated that fish collagen peptide plays a vital role in the growth of selected human skin flora and induces more defensive immunity against pathogenic S. aureus bacteria in dual-culture experimentation.
Collapse
Affiliation(s)
- Rasol Bayan
- Department of Microbiology, Hazara University Mansehra, Mansehra, 21300, Pakistan
| | - Isfahan Tauseef
- Department of Microbiology, Hazara University Mansehra, Mansehra, 21300, Pakistan
| | - Mubashir Hussain
- Department of Microbiology, Kohat University of Science & Technology (KUST), Kohat, 26010, Pakistan
| | - Muhammad S Ahmed
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, 46000, Pakistan
| | - Adnan Haider
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, 46000, Pakistan
| | - Atif Ak Khalil
- Department of Pharmacognosy, Lahore College for Women University, Lahore, 54600, Pakistan
| | - Salman U Islam
- Department of Pharmacy, CECOS University, Peshawar, 25000, Pakistan
| | - Fazli Subhan
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, 46000, Pakistan
| |
Collapse
|
14
|
Mergen B, Onal I, Gulmez A, Caytemel C, Yildirim Y. Conjunctival Microbiota and Blepharitis Symptom Scores in Patients With Ocular Rosacea. Eye Contact Lens 2023; 49:339-343. [PMID: 37363964 DOI: 10.1097/icl.0000000000001008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2023] [Indexed: 06/28/2023]
Abstract
PURPOSE Investigation of the relationship between blepharitis-related symptom scores, tear film functions, and conjunctival microbiota in patients with ocular rosacea (OR) compared with healthy controls was aimed. METHODS Consecutive 33 eyes of 33 patients with OR who were admitted from the dermatology clinic and age-matched and gender-matched 30 healthy controls were included in the study. Tear breakup time (TBUT), Schirmer score, and blepharitis symptom score (BLISS) were recorded for each patient. For the bacteriological examination, bacterial culture was obtained by inoculating the samples on chocolate agar, blood agar, and fluid thioglycollate medium. The growth of different colonies of bacteria was identified using matrix-assisted laser desorption or ionization time-of-flight mass spectrometry. RESULTS Bacterial culture positivity was observed in 13 eyes (39.4%) in the patients with OR and 10 eyes (33.3%) in the controls ( P =0.618). Patients with OR showed worse TBUT and Schirmer scores, and higher BLISSs ( P =0.005, P =0.007, and P =0.001, respectively). Patients with OR with conjunctival culture-positive results showed higher BLISSs (8.0±4.7) compared with those with negative results (4.7±2.3; P <0.001). The most frequent bacteria was Micrococcus luteus (18.2%) in patients with OR and Staphylococcus epidermidis (20.0%) in the controls. CONCLUSION This pilot study showed that patients with OR had similar conjunctival culture positivity compared with healthy controls. However, the observation of different dominant bacterial species in conjunctival microbiota and the observation of worse BLISSs in patients with OR with positive culture might suggest a potential role of conjunctival microbiota in the pathogenesis of OR.
Collapse
Affiliation(s)
- Burak Mergen
- Department of Ophthalmology (B.M., I.O., Y.Y.); Department of Medical Microbiology (A.G.); and Department of Dermatology and Venereology (C.C.), University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | | | | | | | | |
Collapse
|
15
|
Mariani F, Galvan EM. Staphylococcus aureus in Polymicrobial Skinand Soft Tissue Infections: Impact of Inter-Species Interactionsin Disease Outcome. Antibiotics (Basel) 2023; 12:1164. [PMID: 37508260 PMCID: PMC10376372 DOI: 10.3390/antibiotics12071164] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Polymicrobial biofilms provide a complex environment where co-infecting microorganisms can behave antagonistically, additively, or synergistically to alter the disease outcome compared to monomicrobial infections. Staphylococcus aureus skin and soft tissue infections (Sa-SSTIs) are frequently reported in healthcare and community settings, and they can also involve other bacterial and fungal microorganisms. This polymicrobial aetiology is usually found in chronic wounds, such as diabetic foot ulcers, pressure ulcers, and burn wounds, where the establishment of multi-species biofilms in chronic wounds has been extensively described. This review article explores the recent updates on the microorganisms commonly found together with S. aureus in SSTIs, such as Pseudomonas aeruginosa, Escherichia coli, Enterococcus spp., Acinetobacter baumannii, and Candida albicans, among others. The molecular mechanisms behind these polymicrobial interactions in the context of infected wounds and their impact on pathogenesis and antimicrobial susceptibility are also revised.
Collapse
Affiliation(s)
- Florencia Mariani
- Laboratorio de Patogénesis Bacteriana, Departamento de Investigaciones Bioquímicas y Farmacéuticas, Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Hidalgo 775, Buenos Aires C1405, Argentina;
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires A4400, Argentina
| | - Estela Maria Galvan
- Laboratorio de Patogénesis Bacteriana, Departamento de Investigaciones Bioquímicas y Farmacéuticas, Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Hidalgo 775, Buenos Aires C1405, Argentina;
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires A4400, Argentina
| |
Collapse
|
16
|
Wang L, Wang H, Zhang H, Wu H. Formation of a biofilm matrix network shapes polymicrobial interactions. THE ISME JOURNAL 2023; 17:467-477. [PMID: 36639539 PMCID: PMC9938193 DOI: 10.1038/s41396-023-01362-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 12/30/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
Staphylococcus aureus colonizes the same ecological niche as many commensals. However, little is known about how such commensals modulate staphylococcal fitness and persistence. Here we report a new mechanism that mediates dynamic interactions between a commensal streptococcus and S. aureus. Commensal Streptococcus parasanguinis significantly increased the staphylococcal biofilm formation in vitro and enhanced its colonization in vivo. A streptococcal biofilm-associated protein BapA1, not fimbriae-associated protein Fap1, is essential for dual-species biofilm formation. On the other side, three staphylococcal virulence determinants responsible for the BapA1-dependent dual-species biofilm formation were identified by screening a staphylococcal transposon mutant library. The corresponding staphylococcal mutants lacked binding to recombinant BapA1 (rBapA1) due to lower amounts of eDNA in their culture supernatants and were defective in biofilm formation with streptococcus. The rBapA1 selectively colocalized with eDNA within the dual-species biofilm and bound to eDNA in vitro, highlighting the contributions of the biofilm matrix formed between streptococcal BapA1 and staphylococcal eDNA to dual-species biofilm formation. These findings have revealed an additional new mechanism through which an interspecies biofilm matrix network mediates polymicrobial interactions.
Collapse
Affiliation(s)
- Lijun Wang
- Departments of Pediatric Dentistry and Microbiology, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, 35294, USA
- Department of Laboratory Medicine, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, 102218, Beijing, China
| | - Hongxia Wang
- Departments of Pediatric Dentistry and Microbiology, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, 35294, USA
| | - Hua Zhang
- Departments of Pediatric Dentistry and Microbiology, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, 35294, USA
- Department of Integrative Biomedical and Diagnostic Sciences, Oregon Health and Science University School of Dentistry, Portland, OR, 97239, USA
| | - Hui Wu
- Departments of Pediatric Dentistry and Microbiology, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, 35294, USA.
- Department of Integrative Biomedical and Diagnostic Sciences, Oregon Health and Science University School of Dentistry, Portland, OR, 97239, USA.
| |
Collapse
|
17
|
Cui K, Yang W, Liu Z, Liu G, Li D, Sun Y, He G, Ma S, Cao Y, Jiang X, Chevalier S, Cornelis P, Wei Q, Wang Y. Chenodeoxycholic Acid-Amikacin Combination Enhances Eradication of Staphylococcus aureus. Microbiol Spectr 2023; 11:e0243022. [PMID: 36625660 PMCID: PMC9927322 DOI: 10.1128/spectrum.02430-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 12/07/2022] [Indexed: 01/11/2023] Open
Abstract
The rise of antibiotic resistance and dearth of novel antibiotics have posed a serious health crisis worldwide. In this study, we screened a combination of antibiotics and nonantibiotics providing a viable strategy to solve this issue by broadening the antimicrobial spectrum. We found that chenodeoxycholic acid (CDCA), a cholic acid derivative of the traditional Chinese medicine (TCM) Tanreqing (TRQ), synergizes with amikacin against Staphylococcus aureus in vitro, and this synergistic killing was effective against diverse methicillin-resistant S. aureus (MRSA) variants, including small-colony variants (SCVs), biofilm strains, and persisters. The CDCA-amikacin combination protects a mouse model from S. aureus infections. Mechanistically, CDCA increases the uptake of aminoglycosides in a proton motive force-dependent manner by dissipating the chemical potential and potentiates reactive oxygen species (ROS) generation by inhibiting superoxide dismutase activity. This work highlights the potential use of TCM components in treating S. aureus-associated infections and extend the use of aminoglycosides in eradicating Gram-positive pathogens. IMPORTANCE Multidrug resistance (MDR) is spreading globally with increasing speed. The search for new antibiotics is one of the key strategies in the fight against MDR. Antibiotic resistance breakers that may or may not have direct antibacterial action and can either be coadministered or conjugated with other antibiotics are being studied. To better expand the antibacterial spectrum of certain antibiotics, we identified one component from a traditional Chinese medicine, Tanreqing (TRQ), that increased the activity of aminoglycosides. We found that this so-called agent, chenodeoxycholic acid (CDCA), sensitizes Staphylococcus aureus to aminoglycoside killing and protects a mouse model from S. aureus infections. CDCA increases the uptake of aminoglycosides in a proton motive force-dependent manner by dissipating the chemical potential and potentiates ROS generation by inhibiting superoxide dismutase activity in S. aureus. Our work highlights the potential use of TCM or its effective components, such as CDCA, in treating antibiotic resistance-associated infections.
Collapse
Affiliation(s)
- Kaiyu Cui
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Weifeng Yang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhiyuan Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guijian Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dongying Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanan Sun
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Gaiying He
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shuhua Ma
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Cao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuefan Jiang
- Beijing University of Chinese Medicine, Beijing, China
| | - Sylvie Chevalier
- Normandy University, University of Rouen Normandy, Laboratory of Microbiology Signals and Microenvironment, Evreux, France
| | - Pierre Cornelis
- Normandy University, University of Rouen Normandy, Laboratory of Microbiology Signals and Microenvironment, Evreux, France
| | - Qing Wei
- Nanchang Institute of Technology, Nanchang, Jiangxi, China
| | - Yi Wang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
18
|
Pidwill GR, Pyrah JF, Sutton JAF, Best A, Renshaw SA, Foster SJ. Clonal population expansion of Staphylococcus aureus occurs due to escape from a finite number of intraphagocyte niches. Sci Rep 2023; 13:1188. [PMID: 36681703 PMCID: PMC9867732 DOI: 10.1038/s41598-023-27928-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 01/10/2023] [Indexed: 01/22/2023] Open
Abstract
Staphylococcus aureus is a human commensal and also an opportunist pathogen causing life threatening infections. During S. aureus disease, the abscesses that characterise infection can be clonal, whereby a large bacterial population is founded by a single or few organisms. Our previous work has shown that macrophages are responsible for restricting bacterial growth such that a population bottleneck occurs and clonality can emerge. A subset of phagocytes fail to control S. aureus resulting in bacterial division, escape and founding of microabscesses that can seed other host niches. Here we investigate the basis for clonal microabscess formation, using in vitro and in silico models of S. aureus macrophage infection. Macrophages that fail to control S. aureus are characterised by formation of intracellular bacterial masses, followed by cell lysis. High-resolution microscopy reveals that most macrophages had internalised only a single S. aureus, providing a conceptual framework for clonal microabscess generation, which was supported by a stochastic individual-based, mathematical model. Once a threshold of masses was reached, increasing the number of infecting bacteria did not result in greater mass numbers, despite enhanced phagocytosis. This suggests a finite number of permissive, phagocyte niches determined by macrophage associated factors. Increased understanding of the parameters of infection dynamics provides avenues for development of rational control measures.
Collapse
Affiliation(s)
- Grace R Pidwill
- School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK
- Florey Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Josie F Pyrah
- School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK
- Florey Institute, University of Sheffield, Sheffield, S10 2TN, UK
- The Bateson Centre, University of Sheffield, Sheffield, S10 2TN, UK
| | - Joshua A F Sutton
- School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK
- Florey Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Alex Best
- School of Mathematics & Statistics, University of Sheffield, Sheffield, S3 7RH, UK.
| | - Stephen A Renshaw
- Florey Institute, University of Sheffield, Sheffield, S10 2TN, UK.
- The Bateson Centre, University of Sheffield, Sheffield, S10 2TN, UK.
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, S10 2RX, UK.
| | - Simon J Foster
- School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK.
- Florey Institute, University of Sheffield, Sheffield, S10 2TN, UK.
| |
Collapse
|
19
|
Cui K, Yang W, Liu S, Li D, Li L, Ren X, Sun Y, He G, Ma S, Zhang J, Wei Q, Wang Y. Synergistic Inhibition of MRSA by Chenodeoxycholic Acid and Carbapenem Antibiotics. Antibiotics (Basel) 2022; 12:antibiotics12010071. [PMID: 36671273 PMCID: PMC9854648 DOI: 10.3390/antibiotics12010071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 01/03/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) has posed a severe global health threat. In this study, we screened an antibiotic and non-antibiotic combination that provides a viable strategy to solve this issue by broadening the antimicrobial spectrum. We found that chenodeoxycholic acid (CDCA) could synergistically act with carbapenem antibiotics to eradicate MRSA-related infections. This synergy specifically targets MRSA and was also validated using 25 clinical MRSA strains using time-kill analysis. We speculated that the underlying mechanism was associated with the interaction of penicillin-binding proteins (PBPs). As a result, the synergistic efficiency of CDCA with carbapenems targeting PBP1 was better than that of β-lactams targeting PBPs. Moreover, we showed that CDCA did not affect the expression level of PBPs, but sensitized MRSA to carbapenems by disrupting the cell membrane. In our study, we have revealed a novel synergistic combination of antibiotics and non-antibiotics to combat potential bacterial infections.
Collapse
Affiliation(s)
- Kaiyu Cui
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Weifeng Yang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Shuang Liu
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Dongying Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Lu Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xing Ren
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yanan Sun
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Gaiying He
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Shuhua Ma
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jidan Zhang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qing Wei
- Nanchang Institute of Technology, Nanchang 330044, China
- Correspondence: (Q.W.); (Y.W.)
| | - Yi Wang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Correspondence: (Q.W.); (Y.W.)
| |
Collapse
|
20
|
Di Carluccio C, Soriano-Maldonado P, Berni F, de Haas CJC, Temming AR, Hendriks A, Ali S, Molinaro A, Silipo A, van Sorge NM, van Raaij MJ, Codee JDC, Marchetti R. Antibody Recognition of Different Staphylococcus aureus Wall Teichoic Acid Glycoforms. ACS CENTRAL SCIENCE 2022; 8:1383-1392. [PMID: 36313161 PMCID: PMC9615122 DOI: 10.1021/acscentsci.2c00125] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Indexed: 05/14/2023]
Abstract
Wall teichoic acids (WTAs) are glycopolymers decorating the surface of Gram-positive bacteria and potential targets for antibody-mediated treatments against Staphylococcus aureus, including methicillin-resistant (MRSA) strains. Through a combination of glycan microarray, synthetic chemistry, crystallography, NMR, and computational studies, we unraveled the molecular and structural details of fully defined synthetic WTA fragments recognized by previously described monoclonal antibodies (mAbs 4461 and 4497). Our results unveiled the structural requirements for the discriminatory recognition of α- and β-GlcNAc-modified WTA glycoforms by the complementarity-determining regions (CDRs) of the heavy and light chains of the mAbs. Both mAbs interacted not only with the sugar moiety but also with the phosphate groups as well as residues in the ribitol phosphate (RboP) units of the WTA backbone, highlighting their significant role in ligand specificity. Using elongated WTA fragments, containing two sugar modifications, we also demonstrated that the internal carbohydrate moiety of α-GlcNAc-modified WTA is preferentially accommodated in the binding pocket of mAb 4461 with respect to the terminal moiety. Our results also explained the recently documented cross-reactivity of mAb 4497 for β-1,3/β-1,4-GlcNAc-modified WTA, revealing that the flexibility of the RboP backbone is crucial to allow positioning of both glycans in the antibody binding pocket.
Collapse
Affiliation(s)
- Cristina Di Carluccio
- Department
of Chemical Sciences, University of Naples
Federico II, Via Cinthia 4, 80126Naples, Italy
| | - Pablo Soriano-Maldonado
- Departamento
de Estructura de Macromoléculas, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones
Científicas (CNB-CSIC), Calle Darwin 3, 28049Madrid, Spain
| | - Francesca Berni
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CCLeiden, The Netherlands
| | - Carla J. C. de Haas
- Medical
Microbiology, UMC Utrecht, Utrecht University, 3508Utrecht, The Netherlands
| | - A. Robin Temming
- Department
of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, 1105 AZAmsterdam, The Netherlands
| | - Astrid Hendriks
- Department
of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, 1105 AZAmsterdam, The Netherlands
| | - Sara Ali
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CCLeiden, The Netherlands
| | - Antonio Molinaro
- Department
of Chemical Sciences, University of Naples
Federico II, Via Cinthia 4, 80126Naples, Italy
| | - Alba Silipo
- Department
of Chemical Sciences, University of Naples
Federico II, Via Cinthia 4, 80126Naples, Italy
| | - Nina M. van Sorge
- Department
of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, 1105 AZAmsterdam, The Netherlands
- Netherlands
Reference Laboratory for Bacterial Meningitis, Amsterdam UMC, 1105 AZAmsterdam, The Netherlands
- Email
for N.M.v.S.:
| | - Mark J. van Raaij
- Departamento
de Estructura de Macromoléculas, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones
Científicas (CNB-CSIC), Calle Darwin 3, 28049Madrid, Spain
- Email for M.J.v.R.:
| | - Jeroen D. C. Codee
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CCLeiden, The Netherlands
- Email for J.D.C.C.:
| | - Roberta Marchetti
- Department
of Chemical Sciences, University of Naples
Federico II, Via Cinthia 4, 80126Naples, Italy
- Email for R.M.:
| |
Collapse
|
21
|
Rinaldi F, Pinto D, Borsani E, Castrezzati S, Amedei A, Rezzani R. The First Evidence of Bacterial Foci in the Hair Part and Dermal Papilla of Scalp Hair Follicles: A Pilot Comparative Study in Alopecia Areata. Int J Mol Sci 2022; 23:11956. [PMID: 36233254 PMCID: PMC9570265 DOI: 10.3390/ijms231911956] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/29/2022] [Accepted: 10/06/2022] [Indexed: 11/27/2022] Open
Abstract
The role of the microbiome in hair follicle (HF) growth represents a growing field of research. Here, we studied the bacterial population in the scalp hair follicles of subjects with alopecia areata (AA). Two Healthy and two AA subjects, respectively (20−60 years old), were enrolled and studied regarding the microbial community in the subepidermal scalp compartments by means of a 4-mm biopsy punch. Samples were examined by 16S sequencing, histochemical staining (Gram’s method), and transmission electron microscopy (TEM). Bacterial foci were observed in the AA subjects’ follicles with both the two adopted complementary approaches (electron microscopy and Gram staining). Significant (p < 0.05) differences were also found in the three-layer biopsy samples (p < 0.05) regarding the bacterial population. In particular, in the deep epidermis and dermis levels, a significant (p < 0.05) lower abundance of Firmicutes and a higher abundance of Proteobacteria were found in AA samples compared to the healthy control. Firmicutes also showed a significant (p < 0.05) lower abundance in hypodermis in AA subjects. In addition, Enterobacteriaceae and the genera Streptococcus, Gemella, Porphyromonas, and Granulicatella were relatively more abundant in AA groups at the deep epidermis level. The Staphylococcus and Flavobacterium genera were significantly less abundant in AA samples than in controls in all three-layer biopsy samples (p < 0.05). In contrast, Veillonella and Neisseriaceae were relatively more abundant in the healthy control group compared to the AA sample. Therefore, higher alpha diversity was observed in all three-layer biopsy samples of AA patients compared to the control. In conclusion, our data suggest that tAA could be defined as a “hair disease associated with dysregulated microbiome-immunity axis of hair follicles”.
Collapse
Affiliation(s)
- Fabio Rinaldi
- Human Advanced Microbiome Project-HMAP, Giuliani SpA, 20129 Milan, Italy
| | - Daniela Pinto
- Human Advanced Microbiome Project-HMAP, Giuliani SpA, 20129 Milan, Italy
| | - Elisa Borsani
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
- Interdepartmental University Center of Research “Adaption and Regeneration of Tissues and Organs-(ARTO)”, University of Brescia, 25123 Brescia, Italy
| | - Stefania Castrezzati
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Interdisciplinary Internal Medicine Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Rita Rezzani
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy
- Interdepartmental University Center of Research “Adaption and Regeneration of Tissues and Organs-(ARTO)”, University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
22
|
Omaru N, Watanabe T, Kamata K, Minaga K, Kudo M. Activation of NOD1 and NOD2 in the development of liver injury and cancer. Front Immunol 2022; 13:1004439. [PMID: 36268029 PMCID: PMC9577175 DOI: 10.3389/fimmu.2022.1004439] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Hepatocytes and liver-resident antigen-presenting cells are exposed to microbe-associated molecular patterns (MAMPs) and microbial metabolites, which reach the liver from the gut via the portal vein. MAMPs induce innate immune responses via the activation of pattern recognition receptors (PRRs), such as toll-like receptors (TLRs), nucleotide-binding oligomerization domain 1 (NOD1), and NOD2. Such proinflammatory cytokine responses mediated by PRRs likely contribute to the development of chronic liver diseases and hepatocellular carcinoma (HCC), as shown by the fact that activation of TLRs and subsequent production of IL-6 and TNF-α is required for the generation of chronic fibroinflammatory responses and hepatocarcinogenesis. Similar to TLRs, NOD1 and NOD2 recognize MAMPs derived from the intestinal bacteria. The association between the activation of NOD1/NOD2 and chronic liver diseases is poorly understood. Given that NOD1 and NOD2 can regulate proinflammatory cytokine responses mediated by TLRs both positively and negatively, it is likely that sensing of MAMPs by NOD1 and NOD2 affects the development of chronic liver diseases, including HCC. Indeed, recent studies have highlighted the importance of NOD1 and NOD2 activation in chronic liver disorders. Here, we summarize the roles of NOD1 and NOD2 in hepatocarcinogenesis and liver injury.
Collapse
|
23
|
Siwczak F, Cseresnyes Z, Hassan MIA, Aina KO, Carlstedt S, Sigmund A, Groger M, Surewaard BGJ, Werz O, Figge MT, Tuchscherr L, Loffler B, Mosig AS. Human macrophage polarization determines bacterial persistence of Staphylococcus aureus in a liver-on-chip-based infection model. Biomaterials 2022; 287:121632. [PMID: 35728409 DOI: 10.1016/j.biomaterials.2022.121632] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/13/2022] [Accepted: 06/09/2022] [Indexed: 11/17/2022]
Abstract
Infections with Staphylococcus aureus (S. aureus) have been reported from various organs ranging from asymptomatic colonization to severe infections and sepsis. Although considered an extracellular pathogen, S. aureus can invade and persist in professional phagocytes such as monocytes and macrophages. Its capability to persist and manipulate macrophages is considered a critical step to evade host antimicrobial reactions. We leveraged a recently established human liver-on-chip model to demonstrate that S. aureus specifically targets macrophages as essential niche facilitating bacterial persistence and phenotype switching to small colony variants (SCVs). In vitro, M2 polarization was found to favor SCV-formation and was associated with increased intracellular bacterial loads in macrophages, increased cell death, and impaired recruitment of circulating monocytes to sites of infection. These findings expand the knowledge about macrophage activation in the liver and its impact on bacterial persistence and dissemination in the course of infection.
Collapse
Affiliation(s)
- Fatina Siwczak
- Institute of Biochemistry II, Center for Sepsis Control and Care, Jena University Hospital, Am Nonnenplan 1, 07743, Jena, Germany
| | - Zoltan Cseresnyes
- Applied Systems Biology Research Group, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Beutenbergstraße 13, 07745, Jena, Germany
| | - Mohamed I Abdelwahab Hassan
- Institute of Biochemistry II, Center for Sepsis Control and Care, Jena University Hospital, Am Nonnenplan 1, 07743, Jena, Germany
| | - Kehinde Oluwasegun Aina
- Institute of Biochemistry II, Center for Sepsis Control and Care, Jena University Hospital, Am Nonnenplan 1, 07743, Jena, Germany
| | - Swen Carlstedt
- Institute of Biochemistry II, Center for Sepsis Control and Care, Jena University Hospital, Am Nonnenplan 1, 07743, Jena, Germany
| | - Anke Sigmund
- Institute of Medical Microbiology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Marko Groger
- Institute of Biochemistry II, Center for Sepsis Control and Care, Jena University Hospital, Am Nonnenplan 1, 07743, Jena, Germany
| | - Bas G J Surewaard
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, 2500 University Drive NW, Calgary, Alberta, Canada; Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, 3330 Hospital Dr NW, Calgary, Alberta, Canada
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Marc Thilo Figge
- Applied Systems Biology Research Group, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Beutenbergstraße 13, 07745, Jena, Germany; Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, Neugasse 25, 07743, Jena, Germany
| | - Lorena Tuchscherr
- Institute of Medical Microbiology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Bettina Loffler
- Institute of Medical Microbiology, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Alexander S Mosig
- Institute of Biochemistry II, Center for Sepsis Control and Care, Jena University Hospital, Am Nonnenplan 1, 07743, Jena, Germany.
| |
Collapse
|
24
|
Hommes JW, Surewaard BGJ. Intracellular Habitation of Staphylococcus aureus: Molecular Mechanisms and Prospects for Antimicrobial Therapy. Biomedicines 2022; 10:1804. [PMID: 36009351 PMCID: PMC9405036 DOI: 10.3390/biomedicines10081804] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 12/23/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) infections pose a global health threat, especially with the continuous development of antibiotic resistance. As an opportunistic pathogen, MRSA infections have a high mortality rate worldwide. Although classically described as an extracellular pathogen, many studies have shown over the past decades that MRSA also has an intracellular aspect to its infectious cycle, which has been observed in vitro in both non-professional as well as professional phagocytes. In vivo, MRSA has been shown to establish an intracellular niche in liver Kupffer cells upon bloodstream infection. The staphylococci have evolved various evasion strategies to survive the antimicrobial environment of phagolysosomes and use these compartments to hide from immune cells and antibiotics. Ultimately, the host cells get overwhelmed by replicating bacteria, leading to cell lysis and bacterial dissemination. In this review, we describe the different intracellular aspects of MRSA infection and briefly mention S. aureus evasion strategies. We discuss how this intracellular niche of bacteria may assist in antibiotic tolerance development, and lastly, we describe various new antibacterial strategies that target the intracellular bacterial niche.
Collapse
Affiliation(s)
| | - Bas G. J. Surewaard
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada;
| |
Collapse
|
25
|
Fei Y, Ali A, Mohammad M, Jin T. Commensal Bacteria Augment Staphylococcus aureus septic Arthritis in a Dose-Dependent Manner. Front Cell Infect Microbiol 2022; 12:942457. [PMID: 35942056 PMCID: PMC9356218 DOI: 10.3389/fcimb.2022.942457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/23/2022] [Indexed: 12/14/2022] Open
Abstract
Background Septic arthritis is considered one of the most dangerous joints diseases and is mainly caused by the Gram-positive bacterium Staphylococcus aureus (S. aureus). Human skin commensals are known to augment S. aureus infections. The aim of this study was to investigate if human commensals could augment S. aureus-induced septic arthritis. Method NMRI mice were inoculated with S. aureus alone or with a mixture of S. aureus together with either of the human commensal Staphylococcus epidermidis (S. epidermidis) or Streptococcus mitis (S. mitis). The clinical, radiological and histopathological changes due to septic arthritis were observed. Furthermore, the serum levels of chemokines and cytokines were assessed. Results Mice inoculated with a mixture of S. aureus and S. epidermidis or S. mitis developed more severe and frequent clinical arthritis compared to mice inoculated with S. aureus alone. This finding was verified pathologically and radiologically. Furthermore, the ability of mice to clear invading bacteria in the joints but not in kidneys was hampered by the bacterial mixture compared to S. aureus alone. Serum levels of monocyte chemoattractant protein 1 were elevated at the early phase of disease in the mice infected with bacterial mixture compared with ones infected with S. aureus alone. Finally, the augmentation effect in septic arthritis development by S. epidermidis was bacterial dose-dependent. Conclusion The commensal bacteria dose-dependently augment S. aureus-induced septic arthritis in a mouse model of septic arthritis.
Collapse
Affiliation(s)
- Ying Fei
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Microbiology and Immunology, The Affiliated Hospital of GuiZhou Medical University, Guiyang, China
| | - Abukar Ali
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Majd Mohammad
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
- *Correspondence: Tao Jin,
| |
Collapse
|
26
|
Masters EA, Ricciardi BF, Bentley KLDM, Moriarty TF, Schwarz EM, Muthukrishnan G. Skeletal infections: microbial pathogenesis, immunity and clinical management. Nat Rev Microbiol 2022; 20:385-400. [PMID: 35169289 PMCID: PMC8852989 DOI: 10.1038/s41579-022-00686-0] [Citation(s) in RCA: 284] [Impact Index Per Article: 94.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2022] [Indexed: 12/13/2022]
Abstract
Osteomyelitis remains one of the greatest risks in orthopaedic surgery. Although many organisms are linked to skeletal infections, Staphylococcus aureus remains the most prevalent and devastating causative pathogen. Important discoveries have uncovered novel mechanisms of S. aureus pathogenesis and persistence within bone tissue, including implant-associated biofilms, abscesses and invasion of the osteocyte lacuno-canalicular network. However, little clinical progress has been made in the prevention and eradication of skeletal infection as treatment algorithms and outcomes have only incrementally changed over the past half century. In this Review, we discuss the mechanisms of persistence and immune evasion in S. aureus infection of the skeletal system as well as features of other osteomyelitis-causing pathogens in implant-associated and native bone infections. We also describe how the host fails to eradicate bacterial bone infections, and how this new information may lead to the development of novel interventions. Finally, we discuss the clinical management of skeletal infection, including osteomyelitis classification and strategies to treat skeletal infections with emerging technologies that could translate to the clinic in the future.
Collapse
Affiliation(s)
- Elysia A Masters
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
| | - Benjamin F Ricciardi
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Karen L de Mesy Bentley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA.
| | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
27
|
Ndhlovu GON, Dube FS, Moonsamy RT, Mankahla A, Hlela C, Levin ME, Lunjani N, Shittu AO, Abdulgader SM. Skin and nasal colonization of coagulase-negative staphylococci are associated with atopic dermatitis among South African toddlers. PLoS One 2022; 17:e0265326. [PMID: 35298533 PMCID: PMC8929619 DOI: 10.1371/journal.pone.0265326] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 02/28/2022] [Indexed: 11/19/2022] Open
Abstract
Background
Skin colonization with coagulase-negative staphylococci (CoNS) is generally beneficial, but recent investigations suggest its association with flares and atopic dermatitis (AD) severity. However, this relationship remains unclear.
Objective
To assess patterns of staphylococcal colonization and biofilm formation in toddlers with and without AD from rural and urban South African settings.
Methods
We conducted a cross-sectional study of AD-affected and non-atopic AmaXhosa toddlers from rural Umtata and urban Cape Town, South Africa. CoNS isolates were recovered from lesional, nonlesional skin samples and the anterior nares of participants. Identification of the staphylococci was achieved by MALDI-TOF mass spectrometry. The microtiter plate assay assessed in-vitro biofilm formation.
Results
CoNS and S. aureus commonly co-colonized nonlesional skin among cases (urban: 24% vs. 3%, p = 0.037 and rural 21% vs. 6%, p<0.001), and anterior nares in urban cases (24% vs. 0%, p = 0.002) than the control group. S. capitis colonization on nonlesional skin and anterior nares was positively associated with more severe disease in rural (48.3±10.8 vs. 39.7±11.5, P = 0.045) and urban cases (74.9±10.3 vs. 38.4±13, P = 0.004), respectively. Biofilm formation was similar between cases and controls, independent of rural-urban living.
Conclusion
CoNS colonization is associated with AD and disease severity and may be implicated in AD exacerbations. Studies are needed to understand their underlying pathological contribution in AD pathogenesis.
Collapse
Affiliation(s)
- Gillian O. N. Ndhlovu
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Cape Town, South Africa
- * E-mail:
| | - Felix S. Dube
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Rasalika T. Moonsamy
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Avumile Mankahla
- Department of Medicine and Pharmacology, Division of Dermatology, Walter Sisulu University, Umtata, South Africa
| | - Carol Hlela
- Department of Paediatric, Division of Paediatric Allergy, University of Cape Town, Cape Town, South Africa
| | - Michael E. Levin
- Department of Paediatric, Division of Paediatric Allergy, University of Cape Town, Cape Town, South Africa
| | - Nonhlanhla Lunjani
- Department of Paediatric, Division of Paediatric Allergy, University of Cape Town, Cape Town, South Africa
| | - Adebayo O. Shittu
- Department of Microbiology, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Shima M. Abdulgader
- Department of Biomedical Sciences, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
28
|
A. Al-Shammery K, Hozzein WN. Antibacterial activities of two potential peptides extracted from Polistes wattii Cameron, 1900 (Vespidae: Polistinae) wasp venom collected at Eastern Province, Saudi Arabia. PLoS One 2022; 17:e0264035. [PMID: 35255107 PMCID: PMC8901064 DOI: 10.1371/journal.pone.0264035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/02/2022] [Indexed: 11/19/2022] Open
Abstract
Alternatives of conventional antibiotics have become an urgent need to control drug-resistant bacteria. Therefore, search for new antibacterial agents has become a trend in several microbiological and pharmaceutical scientific works. Insects, one of the most successful and evolved species on earth is known to be an effective natural source of several medically useful chemicals including antibacterial agents. There is considerable evidence of using wasp venom against medical ailments in several parts of the world. In this work venom from Polistes wattii Cameron, 1900 collected from Eastern Province, Saudi Arabia was evaluated for its antibacterial activities. Such activity was tested against four pathogenic bacteria: two-gram positive Staphylococcus aureus (ATCC 25923) and Streptococcus mutans (RCMB 017(1) ATCC 25175) and two gram-negative (Salmonella typhimurium NCTC 12023 ATCC 14028 and Enterobacter cloacae (RCMB 001(1) ATCC 23355). Also, chemical characterization of wasp venom was done using HPLC and two isolated peptides were sequenced. The result indicates the potent anti-microbial effect of the venom against the four tested bacteria. The most sensitive bacteria were Staphylococcus aureus (ATCC 25923) and Streptococcus mutans (RCMB 017(1) ATCC 25175). The sequence of the two purified peptides indicates that they belong to mastoparan. The study results may pave way to use this wasp venom in future antibiotics especially in controlling skin infection by Staphylococcus aureus.
Collapse
Affiliation(s)
- Kholoud A. Al-Shammery
- Department of Biology, College of Science, Ha’il University, Ha’il, Saudi Arabia
- * E-mail:
| | - Wael N. Hozzein
- Botany and Microbiology Department, Faculty of Science Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
29
|
Investigating Pathogenicity and Virulence of Staphylococcus pettenkoferi: An Emerging Pathogen. Int J Mol Sci 2021; 22:ijms222413614. [PMID: 34948410 PMCID: PMC8704685 DOI: 10.3390/ijms222413614] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 01/22/2023] Open
Abstract
Staphylococcus pettenkoferi is a coagulase-negative Staphylococcus identified in 2002 that has been implicated in human diseases as an opportunistic pathogenic bacterium. Its multiresistant character is becoming a major health problem, yet the pathogenicity of S. pettenkoferi is poorly characterized. In this study, the pathogenicity of a S. pettenkoferi clinical isolate from diabetic foot osteomyelitis was compared with a Staphylococcus aureus strain in various in vitro and in vivo experiments. Growth kinetics were compared against S. aureus, and bacteria survival was assessed in the RAW 264.7 murine macrophage cell line, the THP-1 human leukemia monocytic cell line, and the HaCaT human keratinocyte cell line. Ex vivo analysis was performed in whole blood survival assays and in vivo assays via the infection model of zebrafish embryos. Moreover, whole-genome analysis was performed. Our results show that S. pettenkoferi was able to survive in human blood, human keratinocytes, murine macrophages, and human macrophages. S. pettenkoferi demonstrated its virulence by causing substantial embryo mortality in the zebrafish model. Genomic analysis revealed virulence factors such as biofilm-encoding genes (e.g., icaABCD; rsbUVW) and regulator-encoding genes (e.g., agr, mgrA, sarA, saeS) well characterized in S. aureus. This study thus advances the knowledge of this under-investigated pathogen and validates the zebrafish infection model for this bacterium.
Collapse
|
30
|
Schoen ME, Jahne MA, Garland J, Ramirez L, Lopatkin AJ, Hamilton KA. Quantitative Microbial Risk Assessment of Antimicrobial Resistant and Susceptible Staphylococcus aureus in Reclaimed Wastewaters. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2021; 55:15246-15255. [PMID: 34699171 PMCID: PMC8721656 DOI: 10.1021/acs.est.1c04038] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The annual risks of colonization, skin infection, bloodstream infection (BSI), and disease burden from exposures to antibiotic-resistant and susceptible Staphylococcus aureus (S. aureus) were estimated using quantitative microbial risk assessment (QMRA). We estimated the probability of nasal colonization after immersion in wastewater (WW) or greywater (GW) treated across a range of treatment alternatives and subsequent infection. Horizontal gene transfer was incorporated into the treatment model but had little effect on the predicted risk. The cumulative annual probability of infection (resulting from self-inoculation) was most sensitive to the treatment log10 reduction value (LRV), S. aureus concentration, and the newly calculated morbidity ratios and was below the health benchmark of 10-4 infections per person per year (ppy) given a treatment LRV of roughly 3.0. The predicted annual disability-adjusted life years (DALYs), which were dominated by BSI, were below the health benchmark of 10-6 DALYs ppy for resistant and susceptible S. aureus, given LRVs of 4.5 and 3.5, respectively. Thus, the estimated infection risks and disease burdens resulting from nasal colonization are below the relevant health benchmarks for risk-based, nonpotable, or potable reuse systems but possibly above for immersion in minimally treated GW or WW. Strain-specific data to characterize dose-response and concentration in WW are needed to substantiate the QMRA.
Collapse
Affiliation(s)
- Mary E Schoen
- Soller Environmental, LLC, 3022 King Street, Berkeley, California 94703, United States
| | - Michael A Jahne
- U.S. Environmental Protection Agency, Office of Research and Development, 26 W. Martin Luther King Drive, Cincinnati, Ohio 45268, United States
| | - Jay Garland
- U.S. Environmental Protection Agency, Office of Research and Development, 26 W. Martin Luther King Drive, Cincinnati, Ohio 45268, United States
| | - Lucia Ramirez
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, Arizona 85281, United States
| | - Allison J Lopatkin
- Department of Biology, Barnard College, New York, New York 10027, United States
- Department Ecology, Evolution, and Environmental Biology, Columbia University, New York, New York 10027, United States
- Data Science Institute, Columbia University, New York, New York 10027, United States
- Department of Systems Biology, Columbia University, New York, New York 10027, United States
| | - Kerry A Hamilton
- School of Sustainable Engineering and the Built Environment, 660 S College Avenue, Tempe Arizona 85281, United States
- The Biodesign Center for Environmental Health Engineering, 1001 S McAllister Avenue, Tempe Arizona 85287, United States
| |
Collapse
|
31
|
MiR-702-3p inhibits the inflammatory injury in septic H9c2 cells by regulating NOD1. Transpl Immunol 2021; 70:101493. [PMID: 34774740 DOI: 10.1016/j.trim.2021.101493] [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: 08/09/2021] [Revised: 10/27/2021] [Accepted: 11/06/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Cardiac insufficiency is a common complication of sepsis and septic shock and is the most common cause of death in critically ill patients. Recent studies have found that microRNAs (miRNAs) play a potential role in sepsis as markers, but little is known about their functional effects on sepsis-induced cardiomyopathy (SIC). OBJECTIVE This study is designed to explore the possible role and underlying mechanisms of miR-702-3p in septic cardiomyopathy. METHODS As expected, H9c2 cells were induced with lipopolysaccharide (LPS) to construct the model of septic cardiomyopathy. The expression of miR-702-3p was detected by qRT-PCR assay and those of IL-1β, IL-6 and TNF-α by ELISA assay. The viability, proliferation and apoptosis of LPS-treated H9c2 cells were determined by CCK-8, EdU, flow cytometry and western blot assays. Moreover, Nucleotide-binding oligomerization domain-containing protein 1 (NOD1) was predicted and confirmed as a direct target of miR-702-3p by TargetScan, miRwalk and miRDB prediction and dual-luciferase reporter gene assays. RESULTS While LPS can weaken the viability of H9c2 cells, miR-702-3p enhances that of LPS-treated H9c2 cells by inhibit the expressions of TNF-α, IL-6, IL-1β. We found NOD1 is a target gene of miR-702-3p, and over-expression of NOD1 restores the inhibitory effects of miR-702-3p on the LPS-treated H9c2 cells. CONCLUSION MiR-702-3p played an important role in the pathogenesis of sepsis cardiomyopathy via targeting NOD1, suggesting that miR-702-3p may be a potential new target for the treatment of SIC.
Collapse
|
32
|
Demonstration of the role of cell wall homeostasis in Staphylococcus aureus growth and the action of bactericidal antibiotics. Proc Natl Acad Sci U S A 2021; 118:2106022118. [PMID: 34716264 PMCID: PMC8612353 DOI: 10.1073/pnas.2106022118] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 09/02/2021] [Indexed: 12/29/2022] Open
Abstract
Bacterial cell wall peptidoglycan is essential, maintaining both cellular integrity and morphology, in the face of internal turgor pressure. Peptidoglycan synthesis is important, as it is targeted by cell wall antibiotics, including methicillin and vancomycin. Here, we have used the major human pathogen Staphylococcus aureus to elucidate both the cell wall dynamic processes essential for growth (life) and the bactericidal effects of cell wall antibiotics (death) based on the principle of coordinated peptidoglycan synthesis and hydrolysis. The death of S. aureus due to depletion of the essential, two-component and positive regulatory system for peptidoglycan hydrolase activity (WalKR) is prevented by addition of otherwise bactericidal cell wall antibiotics, resulting in stasis. In contrast, cell wall antibiotics kill via the activity of peptidoglycan hydrolases in the absence of concomitant synthesis. Both methicillin and vancomycin treatment lead to the appearance of perforating holes throughout the cell wall due to peptidoglycan hydrolases. Methicillin alone also results in plasmolysis and misshapen septa with the involvement of the major peptidoglycan hydrolase Atl, a process that is inhibited by vancomycin. The bactericidal effect of vancomycin involves the peptidoglycan hydrolase SagB. In the presence of cell wall antibiotics, the inhibition of peptidoglycan hydrolase activity using the inhibitor complestatin results in reduced killing, while, conversely, the deregulation of hydrolase activity via loss of wall teichoic acids increases the death rate. For S. aureus, the independent regulation of cell wall synthesis and hydrolysis can lead to cell growth, death, or stasis, with implications for the development of new control regimes for this important pathogen.
Collapse
|
33
|
Bjerre RD, Holm JB, Palleja A, Sølberg J, Skov L, Johansen JD. Skin dysbiosis in the microbiome in atopic dermatitis is site-specific and involves bacteria, fungus and virus. BMC Microbiol 2021; 21:256. [PMID: 34551705 PMCID: PMC8459459 DOI: 10.1186/s12866-021-02302-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 08/28/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Microbial dysbiosis with increased Staphylococcus aureus (S. aureus) colonization on the skin is a hallmark of atopic dermatitis (AD), however most microbiome studies focus on bacteria in the flexures and the microbial composition at other body sites have not been studied systematically. OBJECTIVES The aim of the study is to characterize the skin microbiome, including bacteria, fungi and virus, at different body sites in relation to AD, lesional state, and S. aureus colonization, and to test whether the nares could be a reservoir for S. aureus strain colonization. METHODS Using shotgun metagenomics we characterized microbial compositions from 14 well defined skin sites from 10 patients with AD and 5 healthy controls. RESULTS We found clear differences in microbial composition between AD and controls at multiple skin sites, most pronounced on the flexures and neck. The flexures exhibited lower alpha-diversity and were colonized by S. aureus, accompanied by S. epidermidis in lesions. Malassezia species were absent on the neck in AD. Virus mostly constituted Propionibacterium and Staphylococcus phages, with increased abundance of Propionibacterium phages PHL041 and PHL092 and Staphylococcus epidermidis phages CNPH82 and PH15 in AD. In lesional samples, both the genus Staphylococcus and Staphylococcus phages were more abundant. S. aureus abundance was higher across all skin sites except from the feet. In samples where S. aureus was highly abundant, lower abundances of S. hominis and Cutibacterium acnes were observed. M. osloensis and M. luteus were more abundant in AD. By single nucleotide variant analysis of S. aureus we found strains to be subject specific. On skin sites some S. aureus strains were similar and some dissimilar to the ones in the nares. CONCLUSIONS Our data indicate a global and site-specific dysbiosis in AD, involving both bacteria, fungus and virus. When defining targeted treatment clinicians should both consider the individual and skin site and future research into potential crosstalk between microbiota in AD yields high potential.
Collapse
Affiliation(s)
- Rie Dybboe Bjerre
- National Allergy Research Centre, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark.
| | - Jacob Bak Holm
- Clinical Microbiomics, Fruebjergvej 3, 2100, Copenhagen, Denmark
| | - Albert Palleja
- Clinical Microbiomics, Fruebjergvej 3, 2100, Copenhagen, Denmark
| | - Julie Sølberg
- National Allergy Research Centre, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Lone Skov
- Department of Dermatology and Allergy, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jeanne Duus Johansen
- National Allergy Research Centre, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
34
|
Pandiyan P, McCormick TS. Regulation of IL-17A-Producing Cells in Skin Inflammatory Disorders. J Invest Dermatol 2021; 142:867-875. [PMID: 34561088 DOI: 10.1016/j.jid.2021.06.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/09/2021] [Accepted: 06/22/2021] [Indexed: 12/11/2022]
Abstract
This review focuses on the IL-17A family of cytokines produced by T lymphocytes and other immune cells and how they are involved in cutaneous pathogenic responses. It will also discuss cutaneous dysbiosis and FOXP3+ regulatory T cells in the context of inflammatory conditions linked to IL-17 responses in the skin. Specifically, it will review key literature on chronic mucocutaneous candidiasis and psoriasis.
Collapse
Affiliation(s)
- Pushpa Pandiyan
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.
| | - Thomas S McCormick
- Department of Dermatology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
35
|
Gibson JF, Pidwill GR, Carnell OT, Surewaard BGJ, Shamarina D, Sutton JAF, Jeffery C, Derré-Bobillot A, Archambaud C, Siggins MK, Pollitt EJG, Johnston SA, Serror P, Sriskandan S, Renshaw SA, Foster SJ. Commensal bacteria augment Staphylococcus aureus infection by inactivation of phagocyte-derived reactive oxygen species. PLoS Pathog 2021; 17:e1009880. [PMID: 34529737 PMCID: PMC8478205 DOI: 10.1371/journal.ppat.1009880] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/28/2021] [Accepted: 08/09/2021] [Indexed: 12/22/2022] Open
Abstract
Staphylococcus aureus is a human commensal organism and opportunist pathogen, causing potentially fatal disease. The presence of non-pathogenic microflora or their components, at the point of infection, dramatically increases S. aureus pathogenicity, a process termed augmentation. Augmentation is associated with macrophage interaction but by a hitherto unknown mechanism. Here, we demonstrate a breadth of cross-kingdom microorganisms can augment S. aureus disease and that pathogenesis of Enterococcus faecalis can also be augmented. Co-administration of augmenting material also forms an efficacious vaccine model for S. aureus. In vitro, augmenting material protects S. aureus directly from reactive oxygen species (ROS), which correlates with in vivo studies where augmentation restores full virulence to the ROS-susceptible, attenuated mutant katA ahpC. At the cellular level, augmentation increases bacterial survival within macrophages via amelioration of ROS, leading to proliferation and escape. We have defined the molecular basis for augmentation that represents an important aspect of the initiation of infection. S. aureus is a commensal inhabitant of the human skin and nares. However, it can cause serious diseases if it is able to breach our protective barriers such as the skin, often via wounds or surgery. If infection occurs via a wound, this initial inoculum contains both the pathogen, other members of the microflora and also wider environmental microbes. We have previously described “augmentation”, whereby this other non-pathogenic material can enhance the ability of S. aureus to lead to a serious disease outcome. Here we have determined the breadth of augmenting material and elucidated the cellular and molecular basis for its activity. Augmentation occurs via shielding of S. aureus from the direct bactericidal effects of reactive oxygen species produced by macrophages. This initial protection enables the effective establishment of S. aureus infection. Understanding augmentation not only explains an important facet of the interaction of S. aureus with our innate immune system, but also provides a platform for the development of novel prophylaxis approaches.
Collapse
Affiliation(s)
- Josie F. Gibson
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Grace R. Pidwill
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
| | - Oliver T. Carnell
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
| | - Bas G. J. Surewaard
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Daria Shamarina
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
| | - Joshua A. F. Sutton
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
| | - Charlotte Jeffery
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
| | | | - Cristel Archambaud
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Matthew K. Siggins
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Eric J. G. Pollitt
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
| | - Simon A. Johnston
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Pascale Serror
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Shiranee Sriskandan
- Department of Infectious Disease, Imperial College London, London, United Kingdom
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Stephen A. Renshaw
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular disease, Medical School, University of Sheffield, Sheffield, United Kingdom
- * E-mail: (SAR); (SJF)
| | - Simon J. Foster
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom, Sheffield, United Kingdom
- * E-mail: (SAR); (SJF)
| |
Collapse
|
36
|
Nowik N, Prajsnar TK, Przyborowska A, Rakus K, Sienkiewicz W, Spaink HP, Podlasz P. The Role of Galanin during Bacterial Infection in Larval Zebrafish. Cells 2021; 10:cells10082011. [PMID: 34440783 PMCID: PMC8391356 DOI: 10.3390/cells10082011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/28/2021] [Accepted: 08/03/2021] [Indexed: 11/26/2022] Open
Abstract
Galanin is a peptide that is conserved among different species and plays various roles in an organism, although its entire role is not completely understood. For many years, galanin has been linked mainly with the neurotransmission in the nervous system; however, recent reports underline its role in immunity. Zebrafish (Danio rerio) is an intensively developing animal model to study infectious diseases. In this study, we used larval zebrafish to determine the role of galanin in bacterial infection. We showed that knockout of galanin in zebrafish leads to a higher bacterial burden and mortality during Mycobacterium marinum and Staphylococcus aureus infection, whereas administration of a galanin analogue, NAX 5055, improves the ability of fish to control the infection caused by both pathogens. Moreover, the transcriptomics data revealed that a lower number of genes were regulated in response to mycobacterial infection in gal−/− mutants compared with their gal+/+ wild-type counterparts. We also found that galanin deficiency led to significant changes in immune-related pathways, mostly connected with cytokine and chemokine functions. The results show that galanin acts not only as a neurotransmitter but is also involved in immune response to bacterial infections, demonstrating the complexity of the neuroendocrine system and its possible connection with immunity.
Collapse
Affiliation(s)
- Natalia Nowik
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury, 10-719 Olsztyn, Poland; (N.N.); (A.P.); (W.S.)
- Department of Animal Sciences and Health, Institute of Biology (IBL), Leiden University, 2333 BE Leiden, The Netherlands; (T.K.P.); (H.P.S.)
| | - Tomasz K. Prajsnar
- Department of Animal Sciences and Health, Institute of Biology (IBL), Leiden University, 2333 BE Leiden, The Netherlands; (T.K.P.); (H.P.S.)
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Krakow, Poland;
| | - Anna Przyborowska
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury, 10-719 Olsztyn, Poland; (N.N.); (A.P.); (W.S.)
- Department of Pathophysiology, Forensic Veterinary and Administration, Faculty of Veterinary Medicine, University of Warmia and Mazury, 10-719 Olsztyn, Poland
| | - Krzysztof Rakus
- Department of Evolutionary Immunology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387 Krakow, Poland;
| | - Waldemar Sienkiewicz
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury, 10-719 Olsztyn, Poland; (N.N.); (A.P.); (W.S.)
| | - Herman P. Spaink
- Department of Animal Sciences and Health, Institute of Biology (IBL), Leiden University, 2333 BE Leiden, The Netherlands; (T.K.P.); (H.P.S.)
| | - Piotr Podlasz
- Department of Pathophysiology, Forensic Veterinary and Administration, Faculty of Veterinary Medicine, University of Warmia and Mazury, 10-719 Olsztyn, Poland
- Correspondence: ; Tel.: +48-89-5245291
| |
Collapse
|
37
|
Hommes JW, Kratofil RM, Wahlen S, de Haas CJC, Hildebrand RB, Hovingh GK, Otto M, van Eck M, Hoekstra M, Korporaal SJA, Surewaard BGJ. High density lipoproteins mediate in vivo protection against staphylococcal phenol-soluble modulins. Sci Rep 2021; 11:15357. [PMID: 34321507 PMCID: PMC8319287 DOI: 10.1038/s41598-021-94651-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/28/2021] [Indexed: 12/30/2022] Open
Abstract
Staphylococcus aureus virulence has been associated with the production of phenol-soluble modulins (PSMs). These PSMs have distinct virulence functions and are known to activate, attract and lyse neutrophils. These PSM-associated biological functions are inhibited by lipoproteins in vitro. We set out to address whether lipoproteins neutralize staphylococcal PSM-associated virulence in experimental animal models. Serum from both LCAT an ABCA1 knockout mice strains which are characterised by near absence of high-density lipoprotein (HDL) levels, was shown to fail to protect against PSM-induced neutrophil activation and lysis in vitro. Importantly, PSM-induced peritonitis in LCAT-/- mice resulted in increased lysis of resident peritoneal macrophages and enhanced neutrophil recruitment into the peritoneal cavity. Notably, LCAT-/- mice were more likely to succumb to staphylococcal bloodstream infections in a PSM-dependent manner. Plasma from homozygous carriers of ABCA1 variants characterized by very low HDL-cholesterol levels, was found to be less protective against PSM-mediated biological functions compared to healthy humans. Therefore, we conclude that lipoproteins present in blood can protect against staphylococcal PSMs, the key virulence factor of community-associated methicillin resistant S. aureus.
Collapse
Affiliation(s)
- Josefien W Hommes
- Department of Microbiology, Immunology, and Infectious Disease. Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Rachel M Kratofil
- Department of Microbiology, Immunology, and Infectious Disease. Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Sigrid Wahlen
- Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Diagnostic Sciences, Laboratory of Experimental Immunology, Ghent University, Ghent, Belgium
| | - Carla J C de Haas
- Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Reeni B Hildebrand
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Leiden, The Netherlands
| | - G Kees Hovingh
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Micheal Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Miranda van Eck
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Leiden, The Netherlands
| | - Menno Hoekstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Leiden, The Netherlands
| | - Suzanne J A Korporaal
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Gorlaeus Laboratories, Leiden, The Netherlands.,Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bas G J Surewaard
- Department of Microbiology, Immunology, and Infectious Disease. Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada. .,Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
38
|
A large epidemic of a necrotic skin infection in the Democratic Republic of São Tomé and Principe: an epidemiological study. Int J Infect Dis 2021; 110 Suppl 1:S69-S76. [PMID: 34246783 DOI: 10.1016/j.ijid.2021.06.050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION In 2016-18, the Democratic Republic of São Tomé and Príncipe suffered a necrotic skin infection epidemic. METHODS A surveillance system was established after increased hospitalisations for this infection. Microbiology results were available for samples analysed in December 2016 and March 2017 using whole genome sequencing and metagenomics. Negative binomial regression was used to study the association of weather conditions with monthly case counts in a time-series analysis. RESULTS From October 2016 to October 2018, the epidemic cumulative attack rate was 1.5%. The first peak lasted 5 months, accounting for one-third of total cases. We could not conclusively identify the aetiological agent(s) due to the country's lack of microbiology capacity. Increased relative humidity was associated with increased monthly cases (incidence rate ratio (IRR) 1.05, 95% CI 1.02-1.09), and higher precipitation in the previous month with a higher number of cases in the following month (months with 0-49 mm rainfall compared with months with 50-149 mm and ≥150 mm: IRR 1.44, 95 % CI 1.13-1.78 and 1.50, 95% CI 1.12-1.99, respectively). DISCUSSION This epidemic was favoured by increased relative humidity and precipitation, potentially contributing to community-based transmission of ubiquitous bacterial strains superinfecting skin wounds. FUNDING World Health Organization Regional Office for Africa, Ministry of Health.
Collapse
|
39
|
Rasheed S, Fries F, Müller R, Herrmann J. Zebrafish: An Attractive Model to Study Staphylococcus aureus Infection and Its Use as a Drug Discovery Tool. Pharmaceuticals (Basel) 2021; 14:594. [PMID: 34205723 PMCID: PMC8235121 DOI: 10.3390/ph14060594] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 11/16/2022] Open
Abstract
Non-mammalian in vivo disease models are particularly popular in early drug discovery. Zebrafish (Danio rerio) is an attractive vertebrate model, the success of which is driven by several advantages, such as the optical transparency of larvae, the small and completely sequenced genome, the small size of embryos and larvae enabling high-throughput screening, and low costs. In this review, we highlight zebrafish models of Staphyloccoccus aureus infection, which are used in drug discovery and for studying disease pathogenesis and virulence. Further, these infection models are discussed in the context of other relevant zebrafish models for pharmacological and toxicological studies as part of early drug profiling. In addition, we examine key differences to commonly applied models of S.aureus infection based on invertebrate organisms, and we compare their frequency of use in academic research covering the period of January 2011 to January 2021.
Collapse
Affiliation(s)
- Sari Rasheed
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123 Saarbrücken, Germany; (S.R.); (F.F.); (R.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover–Braunschweig, 38124 Braunschweig, Germany
| | - Franziska Fries
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123 Saarbrücken, Germany; (S.R.); (F.F.); (R.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover–Braunschweig, 38124 Braunschweig, Germany
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123 Saarbrücken, Germany; (S.R.); (F.F.); (R.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover–Braunschweig, 38124 Braunschweig, Germany
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Jennifer Herrmann
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Saarland University Campus, 66123 Saarbrücken, Germany; (S.R.); (F.F.); (R.M.)
- German Centre for Infection Research (DZIF), Partner Site Hannover–Braunschweig, 38124 Braunschweig, Germany
| |
Collapse
|
40
|
Synergistic In Vitro Antimicrobial Activity of Pomegranate Rind Extract and Zinc (II) against Micrococcus luteus under Planktonic and Biofilm Conditions. Pharmaceutics 2021; 13:pharmaceutics13060851. [PMID: 34201223 PMCID: PMC8230037 DOI: 10.3390/pharmaceutics13060851] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/24/2021] [Accepted: 05/31/2021] [Indexed: 01/02/2023] Open
Abstract
Infectious diseases caused by microbial biofilms are a major clinical problem, and new antimicrobial agents that can inhibit biofilm formation and eradicate pre-formed biofilms are urgently needed. Pomegranate extracts are a well-established folkloric medicine and have been used in the treatment of infectious diseases since ancient times, whilst the addition of metal ions, including zinc (II), has enhanced the antimicrobial activity of pomegranate. Micrococcus luteus is generally a non-pathogenic skin commensal bacterium, although it can act as an opportunistic pathogen and cause serious infections, particularly involving catheterization and comorbidities. The aims of this study were to evaluate the holistic activity of pomegranate rind extract (PRE), Zn (II), and PRE/Zn (II) individually and in combination against M. luteus under both planktonic and biofilm conditions. Antimicrobial activity was detected in vitro using the broth dilution method, and synergistic activity was determined using checkerboard and time-kill assays. Effects on biofilm formation and eradication were determined by crystal violet and BacLightTM Live/Dead staining. PRE and Zn (II) exerted antimicrobial activity against M. luteus under both planktonic and biofilm conditions. After 4 h, potent synergistic bactericidal activity was also found when PRE and Zn (II) were co-administered under planktonic conditions (log reductions: PRE 1.83 ± 0.24, Zn (II) 3.4 ± 0.08, and PRE/Zn (II) 6.88 ± 1.02; p < 0.0001). In addition, greater heterogeneity was induced in the structure of M. luteus biofilm using the PRE/Zn (II) combination compared to when PRE and Zn (II) were applied individually. The activity of PRE and the PRE/Zn (II) combination could offer a novel antimicrobial therapy for the treatment of disease-associated infections caused by M. luteus and potentially other bacteria.
Collapse
|
41
|
Gibson JF, Prajsnar TK, Hill CJ, Tooke AK, Serba JJ, Tonge RD, Foster SJ, Grierson AJ, Ingham PW, Renshaw SA, Johnston SA. Neutrophils use selective autophagy receptor Sqstm1/p62 to target Staphylococcus aureus for degradation in vivo in zebrafish. Autophagy 2021; 17:1448-1457. [PMID: 32559122 PMCID: PMC8204994 DOI: 10.1080/15548627.2020.1765521] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 04/24/2020] [Accepted: 05/02/2020] [Indexed: 12/19/2022] Open
Abstract
Macroautophagy/autophagy functions to degrade cellular components and intracellular pathogens. Autophagy receptors, including SQSTM1/p62, target intracellular pathogens. Staphylococcus aureus is a significant pathogen of humans, especially in immunocompromise. S. aureus may use neutrophils as a proliferative niche, but their intracellular fate following phagocytosis has not been analyzed in vivo. In vitro, SQSTM1 can colocalize with intracellular Staphylococcus aureus, but whether SQSTM1 is beneficial or detrimental in host defense against S. aureus in vivo is unknown. Here we determine the fate and location of S. aureus within neutrophils throughout zebrafish infection. We show Lc3 and Sqstm1 recruitment to phagocytosed S. aureus is altered depending on the bacterial location within the neutrophil and that Lc3 marking of bacterial phagosomes within neutrophils may precede bacterial degradation. Finally, we show Sqstm1 is important for controlling cytosolic bacteria, demonstrating for the first time a key role of Sqstm1 in autophagic control of S. aureus in neutrophils.Abbreviations: AR: autophagy receptor; CFU: colony-forming unit; CHT: caudal hematopoietic tissue; GFP: green fluorescent protein; hpf: hours post-fertilization; hpi: hours post-infection; LWT: london wild-type: lyz: lysozyme; Map1lc3/Lc3: microtubule-associated protein 1 light chain 3; RFP: red fluorescent protein; Sqstm1/p62: sequestosome 1; Tg: transgenic; TSA: tyramide signal amplification; UBD: ubiquitin binding domain.
Collapse
Affiliation(s)
- Josie F. Gibson
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK
- The Bateson Centre, University of Sheffield, Sheffield, UK
- Institute of Molecular and Cell Biology, Agency of Science, Technology and Research (A-star), Singapore
- Florey Institute, University of Sheffield, Sheffield, UK
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, UK
| | - Tomasz K. Prajsnar
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK
- The Bateson Centre, University of Sheffield, Sheffield, UK
- Institute Biology Leiden, Leiden University, Leiden, The Netherlands
| | - Christopher J. Hill
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, UK
| | - Amy K. Tooke
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, UK
| | - Justyna J. Serba
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK
- The Bateson Centre, University of Sheffield, Sheffield, UK
| | - Rebecca D. Tonge
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield, UK
| | - Simon J Foster
- Florey Institute, University of Sheffield, Sheffield, UK
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, UK
| | - Andrew J. Grierson
- The Bateson Centre, University of Sheffield, Sheffield, UK
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield, UK
| | - Philip W. Ingham
- Institute of Molecular and Cell Biology, Agency of Science, Technology and Research (A-star), Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Stephen A. Renshaw
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK
- The Bateson Centre, University of Sheffield, Sheffield, UK
- Florey Institute, University of Sheffield, Sheffield, UK
| | - Simon A. Johnston
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK
- The Bateson Centre, University of Sheffield, Sheffield, UK
- Florey Institute, University of Sheffield, Sheffield, UK
| |
Collapse
|
42
|
Abstract
The skin microbiome is an ecosystem comprised of a multitude of microbial species interacting with their surroundings, including other microbes and host epithelial and immune cells. These interactions are the basis of important roles within the skin microbiome that provide benefit to the host, boosting multiple aspects of barrier function, a critical function of this essential organ. However, with reward always comes risk; resident skin microbes function in a context-dependent manner, set on the backdrop of a dynamic host and microbial milieu. Here, we discuss the reward of hosting a microbial ecosystem on the skin, including protection from pathogens and tuning of the skin microenvironment. We also give consideration to how these skin residents, often termed "commensals" can cause disorder, damage, and promote skin disease.
Collapse
Affiliation(s)
- Laurice Flowers
- Department of Dermatology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Elizabeth A Grice
- Department of Dermatology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
43
|
The Ambivalent Role of Skin Microbiota and Adrenaline in Wound Healing and the Interplay between Them. Int J Mol Sci 2021; 22:ijms22094996. [PMID: 34066786 PMCID: PMC8125934 DOI: 10.3390/ijms22094996] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 04/27/2021] [Accepted: 05/06/2021] [Indexed: 02/07/2023] Open
Abstract
After skin injury, wound healing sets into motion a dynamic process to repair and replace devitalized tissues. The healing process can be divided into four overlapping phases: hemostasis, inflammation, proliferation, and maturation. Skin microbiota has been reported to participate in orchestrating the wound healing both in negative and positive ways. Many studies reported that skin microbiota can impose negative and positive effects on the wound. Recent findings have shown that many bacterial species on human skin are able to convert aromatic amino acids into so-called trace amines (TAs) and convert corresponding precursors into dopamine and serotonin, which are all released into the environment. As a stress reaction, wounded epithelial cells release the hormone adrenaline (epinephrine), which activates the β2-adrenergic receptor (β2-AR), impairing the migration ability of keratinocytes and thus re-epithelization. This is where TAs come into play, as they act as antagonists of β2-AR and thus attenuate the effects of adrenaline. The result is that not only TAs but also TA-producing skin bacteria accelerate wound healing. Adrenergic receptors (ARs) play a key role in many physiological and disease-related processes and are expressed in numerous cell types. In this review, we describe the role of ARs in relation to wound healing in keratinocytes, immune cells, fibroblasts, and blood vessels and the possible role of the skin microbiota in wound healing.
Collapse
|
44
|
Prajsnar TK, Serba JJ, Dekker BM, Gibson JF, Masud S, Fleming A, Johnston SA, Renshaw SA, Meijer AH. The autophagic response to Staphylococcus aureus provides an intracellular niche in neutrophils. Autophagy 2021; 17:888-902. [PMID: 32174246 PMCID: PMC8078660 DOI: 10.1080/15548627.2020.1739443] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 02/17/2020] [Accepted: 02/28/2020] [Indexed: 11/22/2022] Open
Abstract
Staphylococcus aureus is a major human pathogen causing multiple pathologies, from cutaneous lesions to life-threatening sepsis. Although neutrophils contribute to immunity against S. aureus, multiple lines of evidence suggest that these phagocytes can provide an intracellular niche for staphylococcal dissemination. However, the mechanism of neutrophil subversion by intracellular S. aureus remains unknown. Targeting of intracellular pathogens by macroautophagy/autophagy is recognized as an important component of host innate immunity, but whether autophagy is beneficial or detrimental to S. aureus-infected hosts remains controversial. Here, using larval zebrafish, we showed that the autophagy marker Lc3 rapidly decorates S. aureus following engulfment by macrophages and neutrophils. Upon phagocytosis by neutrophils, Lc3-positive, non-acidified spacious phagosomes are formed. This response is dependent on phagocyte NADPH oxidase as both cyba/p22phox knockdown and diphenyleneiodonium (DPI) treatment inhibited Lc3 decoration of phagosomes. Importantly, NADPH oxidase inhibition diverted neutrophil S. aureus processing into tight acidified vesicles, which resulted in increased host resistance to the infection. Some intracellular bacteria within neutrophils were also tagged by Sqstm1/p62-GFP fusion protein and loss of Sqstm1 impaired host defense. Together, we have shown that intracellular handling of S. aureus by neutrophils is best explained by Lc3-associated phagocytosis (LAP), which appears to provide an intracellular niche for bacterial pathogenesis, while the selective autophagy receptor Sqstm1 is host-protective. The antagonistic roles of LAP and Sqstm1-mediated pathways in S. aureus-infected neutrophils may explain the conflicting reports relating to anti-staphylococcal autophagy and provide new insights for therapeutic strategies against antimicrobial-resistant Staphylococci.Abbreviations: ATG: autophagy related; CFU: colony-forming units; CMV: cytomegalovirus; Cyba/P22phox: cytochrome b-245, alpha polypeptide; DMSO: dimethyl sulfoxide; DPI: diphenyleneiodonium; EGFP: enhanced green fluorescent protein; GFP: green fluorescent protein; hpf: hours post-fertilization; hpi: hours post-infection; Irf8: interferon regulatory factor 8; LAP: LC3-associated phagocytosis; lyz: lysozyme; LWT: london wild type; Map1lc3/Lc3: microtubule-associated protein 1 light chain 3; NADPH oxidase: nicotinamide adenine dinucleotide phosphate oxidase; RFP: red fluorescent protein; ROS: reactive oxygen species; RT-PCR: reverse transcriptase polymerase chain reaction; Sqstm1/p62: sequestosome 1; Tg: transgenic; TSA: tyramide signal amplification.
Collapse
Affiliation(s)
- Tomasz K. Prajsnar
- Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Institute of Biology Leiden, Faculty of Science, Leiden University, Leiden, The Netherlands
| | - Justyna J. Serba
- Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Bernice M. Dekker
- Institute of Biology Leiden, Faculty of Science, Leiden University, Leiden, The Netherlands
| | - Josie F. Gibson
- Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Krebs Institute and Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, UK
| | - Samrah Masud
- Institute of Biology Leiden, Faculty of Science, Leiden University, Leiden, The Netherlands
| | - Angeleen Fleming
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Simon A. Johnston
- Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Stephen A. Renshaw
- Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Annemarie H. Meijer
- Institute of Biology Leiden, Faculty of Science, Leiden University, Leiden, The Netherlands
| |
Collapse
|
45
|
Staphylococcus aureus cell wall structure and dynamics during host-pathogen interaction. PLoS Pathog 2021; 17:e1009468. [PMID: 33788901 PMCID: PMC8041196 DOI: 10.1371/journal.ppat.1009468] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/12/2021] [Accepted: 03/12/2021] [Indexed: 01/09/2023] Open
Abstract
Peptidoglycan is the major structural component of the Staphylococcus aureus cell wall, in which it maintains cellular integrity, is the interface with the host, and its synthesis is targeted by some of the most crucial antibiotics developed. Despite this importance, and the wealth of data from in vitro studies, we do not understand the structure and dynamics of peptidoglycan during infection. In this study we have developed methods to harvest bacteria from an active infection in order to purify cell walls for biochemical analysis ex vivo. Isolated ex vivo bacterial cells are smaller than those actively growing in vitro, with thickened cell walls and reduced peptidoglycan crosslinking, similar to that of stationary phase cells. These features suggested a role for specific peptidoglycan homeostatic mechanisms in disease. As S. aureus missing penicillin binding protein 4 (PBP4) has reduced peptidoglycan crosslinking in vitro its role during infection was established. Loss of PBP4 resulted in an increased recovery of S. aureus from the livers of infected mice, which coincided with enhanced fitness within murine and human macrophages. Thicker cell walls correlate with reduced activity of peptidoglycan hydrolases. S. aureus has a family of 4 putative glucosaminidases, that are collectively crucial for growth. Loss of the major enzyme SagB, led to attenuation during murine infection and reduced survival in human macrophages. However, loss of the other three enzymes Atl, SagA and ScaH resulted in clustering dependent attenuation, in a zebrafish embryo, but not a murine, model of infection. A combination of pbp4 and sagB deficiencies resulted in a restoration of parental virulence. Our results, demonstrate the importance of appropriate cell wall structure and dynamics during pathogenesis, providing new insight to the mechanisms of disease. The prevalence of methicillin resistant Staphylococcus aureus (MRSA) in both hospitals and the wider community places a huge weight on healthcare providers. To discover new control regimes, it is therefore important to understand how the pathogen behaves within the relevant environment of the host. This is often hampered by the ability to obtain sufficient ex vivo pathogen samples for study. We have developed a method to isolate S. aureus from the infected host to be able to analyse cellular morphology and structure. S. aureus, isolated from an infected kidney abscess are smaller in size, with thicker cell walls than exponentially growing cells in vitro. Their cell wall peptidoglycan also is less crosslinked. These features suggested the role of components controlling cell wall homeostasis as being important for infections. We tested the role of PBP4, known to increase cell wall crosslinking and found a pbp4 mutant to have increased survival in macrophages and fitness within the murine host. Conversely the peptidoglycan hydrolase SagB, whose loss results in thinner cell walls was attenuated in the murine systemic model of infection, with concomitant loss of fitness within macrophages. Our study reveals an important adaptation to the host environment and the role of those components involved in cell wall homeostasis in vivo.
Collapse
|
46
|
Mohammad M, Na M, Hu Z, Nguyen MT, Kopparapu PK, Jarneborn A, Karlsson A, Ali A, Pullerits R, Götz F, Jin T. Staphylococcus aureus lipoproteins promote abscess formation in mice, shielding bacteria from immune killing. Commun Biol 2021; 4:432. [PMID: 33785850 PMCID: PMC8010101 DOI: 10.1038/s42003-021-01947-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 03/02/2021] [Indexed: 12/27/2022] Open
Abstract
Despite being a major bacterial factor in alerting the human immune system, the role of Staphylococcus aureus (S. aureus) lipoproteins (Lpp) in skin infections remains largely unknown. Here, we demonstrated that subcutaneous injection of S. aureus Lpp led to infiltration of neutrophils and monocytes/macrophages and induced skin lesions in mice. Lipid-moiety of S. aureus Lpp and host TLR2 was responsible for such effect. Lpp-deficient S. aureus strains exhibited smaller lesion size and reduced bacterial loads than their parental strains; the altered phenotype in bacterial loads was TLR2-independent. Lpp expression in skin infections contributed to imbalanced local hemostasis toward hypercoagulable state. Depletion of leukocytes or fibrinogen abrogated the effects induced by Lpp in terms of skin lesions and bacterial burden. Our data suggest that S. aureus Lpp induce skin inflammation and promote abscess formation that protects bacteria from innate immune killing. This suggests an intriguing bacterial immune evasion mechanism.
Collapse
Affiliation(s)
- Majd Mohammad
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Manli Na
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Zhicheng Hu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Microbiology and Immunology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Minh-Thu Nguyen
- Department of Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Tübingen, Germany
- Section of Medical and Geographical Infectiology, Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Pradeep Kumar Kopparapu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders Jarneborn
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anna Karlsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Abukar Ali
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Rille Pullerits
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Friedrich Götz
- Department of Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Tübingen, Germany
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
47
|
Analysis of Staphylococcus aureus Transcriptome in Pediatric Soft Tissue Abscesses and Comparison to Murine Infections. Infect Immun 2021; 89:IAI.00715-20. [PMID: 33526560 DOI: 10.1128/iai.00715-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 01/17/2023] Open
Abstract
A comprehensive understanding of how Staphylococcus aureus adapts to cause infections in humans can inform development of diagnostic, therapeutic, and preventive approaches. Expression analysis of clinical strain libraries depicts in vitro conditions that differ from those in human infection, but low bacterial burden and the requirement for reverse transcription or nucleic acid amplification complicate such analyses of bacteria causing human infection. We developed methods to evaluate the mRNA transcript signature of S. aureus in pediatric skin and soft tissue infections (SSTI) directly ex vivo Abscess drainage from 47 healthy pediatric patients undergoing drainage of a soft tissue infection was collected, and RNA was extracted from samples from patients with microbiologically confirmed S. aureus abscesses (42% due to methicillin-resistant S. aureus [MRSA]). Using the NanoString platform and primers targeting S. aureus mRNA transcripts encoding surface-expressed or secreted proteins, we measured direct counts of 188 S. aureus mRNA transcripts in abscess drainage. We further evaluated this mRNA signature in murine models of S. aureus SSTI and nasal colonization where the kinetics of the transcriptome could be determined. Heat maps of the S. aureus mRNA signatures from pediatric abscesses demonstrated consistent per-target expression across patients. While there was significant overlap with the profiles from murine SSTI and nasal colonization, important differences were noted, which can inform efforts to develop therapeutic and vaccine approaches.
Collapse
|
48
|
Soe YM, Bedoui S, Stinear TP, Hachani A. Intracellular Staphylococcus aureus and host cell death pathways. Cell Microbiol 2021; 23:e13317. [PMID: 33550697 DOI: 10.1111/cmi.13317] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/22/2021] [Accepted: 01/23/2021] [Indexed: 12/13/2022]
Abstract
Staphylococcus aureus is a major opportunistic human pathogen that is globally prevalent. Although S. aureus and humans may have co-evolved to the point of commensalism, the bacterium is equipped with virulence factors causing devastating infections. The adoption of an intracellular lifestyle by S. aureus is an important facet of its pathogenesis. Occupying a privileged intracellular compartment permits evasion from the bactericidal actions of host immunity and antibiotics. However, this localization exposes S. aureus to cell-intrinsic processes comprising autophagy, metabolic challenges and clearance mechanisms orchestrated by host programmed cell death pathways (PCDs), including apoptosis, pyroptosis and necroptosis. Mounting evidence suggests that S. aureus deploys pathoadaptive mechanisms that modulate the expression of its virulence factors to prevent elimination through PCD pathways. In this review, we critically analyse the current literature on the interplay between S. aureus virulence factors with the key, intertwined nodes of PCD. We discuss how S. aureus adaptation to the human host plays an essential role in the evasion of PCD, and we consider future directions to study S. aureus-PCD interactions.
Collapse
Affiliation(s)
- Ye Mon Soe
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Sammy Bedoui
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Timothy P Stinear
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Abderrahman Hachani
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
49
|
Pidwill GR, Gibson JF, Cole J, Renshaw SA, Foster SJ. The Role of Macrophages in Staphylococcus aureus Infection. Front Immunol 2021; 11:620339. [PMID: 33542723 PMCID: PMC7850989 DOI: 10.3389/fimmu.2020.620339] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/02/2020] [Indexed: 12/23/2022] Open
Abstract
Staphylococcus aureus is a member of the human commensal microflora that exists, apparently benignly, at multiple sites on the host. However, as an opportunist pathogen it can also cause a range of serious diseases. This requires an ability to circumvent the innate immune system to establish an infection. Professional phagocytes, primarily macrophages and neutrophils, are key innate immune cells which interact with S. aureus, acting as gatekeepers to contain and resolve infection. Recent studies have highlighted the important roles of macrophages during S. aureus infections, using a wide array of killing mechanisms. In defense, S. aureus has evolved multiple strategies to survive within, manipulate and escape from macrophages, allowing them to not only subvert but also exploit this key element of our immune system. Macrophage-S. aureus interactions are multifaceted and have direct roles in infection outcome. In depth understanding of these host-pathogen interactions may be useful for future therapeutic developments. This review examines macrophage interactions with S. aureus throughout all stages of infection, with special emphasis on mechanisms that determine infection outcome.
Collapse
Affiliation(s)
- Grace R. Pidwill
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
| | - Josie F. Gibson
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Joby Cole
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Stephen A. Renshaw
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Simon J. Foster
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
50
|
Gonzalez T, Stevens ML, Baatyrbek kyzy A, Alarcon R, He H, Kroner JW, Spagna D, Grashel B, Sidler E, Martin LJ, Biagini Myers JM, Khurana Hershey GK, Herr AB. Biofilm propensity of Staphylococcus aureus skin isolates is associated with increased atopic dermatitis severity and barrier dysfunction in the MPAACH pediatric cohort. Allergy 2021; 76:302-313. [PMID: 32640045 DOI: 10.1111/all.14489] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 04/30/2020] [Accepted: 05/24/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Atopic dermatitis (AD) patients are often colonized with Staphylococcus aureus, and staphylococcal biofilms have been reported on adult AD skin lesions. The commensal S epidermidis can antagonize S aureus, although its role in AD is unclear. We sought to characterize S aureus and S epidermidis colonization and biofilm propensity and determine their associations with AD severity, barrier function, and epidermal gene expression in the first US early-life cohort of children with AD, the Mechanisms of Progression of Atopic Dermatitis to Asthma in Children (MPAACH). METHODS The biofilm propensity of staphylococcal isolates was assessed by crystal violet assays. Gene expression of filaggrin and antimicrobial alarmins S100A8 and S100A9 was measured in keratinocyte RNA extracted from skin tape strips. Staphylococcal biofilms sampled from MPAACH skin were visualized using scanning electron microscopy. RESULTS Sixty-two percent of staphylococcal isolates (sampled from 400 subjects) formed moderate/strong biofilms. Sixty-eight percent of subjects co-colonized with both staphylococcal species exhibited strains that formed cooperative mixed-species biofilms. Scanning electron microscopy verified the presence of staphylococcal biofilms on the skin of MPAACH children. Staphylococcus aureus strains showing higher relative biofilm propensity compared with S epidermidis were associated with increased AD severity (P = .03) and increased lesional and nonlesional transepidermal water loss (P = .01, P = .03). CONCLUSIONS Our data suggest a pathogenic role for S aureus biofilms in AD. We found that strain-level variation in staphylococcal isolates governs the interactions between S epidermidis and S aureus and that the balance between these two species, and their biofilm propensity, has important implications for AD.
Collapse
Affiliation(s)
- Tammy Gonzalez
- Division of Immunobiology Cincinnati Children's Hospital Medical Center Cincinnati OH USA
| | - Mariana L. Stevens
- Division of Asthma Research Cincinnati Children's Hospital Medical Center Cincinnati OH USA
| | - Asel Baatyrbek kyzy
- Division of Asthma Research Cincinnati Children's Hospital Medical Center Cincinnati OH USA
| | - Rosario Alarcon
- Division of Asthma Research Cincinnati Children's Hospital Medical Center Cincinnati OH USA
| | - Hua He
- Division of Human Genetics Cincinnati Children's Hospital Medical Center Cincinnati OH USA
| | - John W. Kroner
- Division of Asthma Research Cincinnati Children's Hospital Medical Center Cincinnati OH USA
| | - Daniel Spagna
- Division of Asthma Research Cincinnati Children's Hospital Medical Center Cincinnati OH USA
| | - Brittany Grashel
- Division of Asthma Research Cincinnati Children's Hospital Medical Center Cincinnati OH USA
| | - Elaine Sidler
- Division of Immunobiology Cincinnati Children's Hospital Medical Center Cincinnati OH USA
| | - Lisa J. Martin
- Division of Human Genetics Cincinnati Children's Hospital Medical Center Cincinnati OH USA
- Department of Pediatrics University of Cincinnati College of Medicine Cincinnati OH USA
| | - Jocelyn M. Biagini Myers
- Division of Asthma Research Cincinnati Children's Hospital Medical Center Cincinnati OH USA
- Department of Pediatrics University of Cincinnati College of Medicine Cincinnati OH USA
| | - Gurjit K. Khurana Hershey
- Division of Asthma Research Cincinnati Children's Hospital Medical Center Cincinnati OH USA
- Department of Pediatrics University of Cincinnati College of Medicine Cincinnati OH USA
- Division of Allergy and Immunology Cincinnati Children's Hospital Medical Center Cincinnati OH USA
| | - Andrew B. Herr
- Division of Immunobiology Cincinnati Children's Hospital Medical Center Cincinnati OH USA
- Department of Pediatrics University of Cincinnati College of Medicine Cincinnati OH USA
- Division of Infectious Diseases Cincinnati Children's Hospital Medical Center Cincinnati OH USA
| |
Collapse
|