1
|
Østerlid KE, Sorieul C, Unione L, Li S, García-Sepúlveda C, Carboni F, Del Bino L, Berni F, Arda A, Overkleeft HS, van der Marel GA, Romano MR, Jiménez-Barbero J, Adamo R, Codée JDC. Long, Synthetic Staphylococcus aureus Type 8 Capsular Oligosaccharides Reveal Structural Epitopes for Effective Immune Recognition. J Am Chem Soc 2025; 147:2829-2840. [PMID: 39792791 PMCID: PMC11760181 DOI: 10.1021/jacs.4c16118] [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: 11/15/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Staphylococcus aureus is a Gram-positive bacterium that is responsible for severe nosocomial infections. The rise of multidrug-resistant strains, which can pose significant health threats, prompts the development of new treatment interventions, and much attention has been directed at the development of prophylactic and therapeutic vaccination strategies. Capsular polysaccharides (CPs) are key protective elements of the S. aureus cell wall and have been proposed as promising candidate antigens. Thirteen different CP serotypes have been identified to date, of which types 5 and 8 are the most prominent. CP8 is composed of trisaccharide repeating units that are built up from an N-acetyl-4-O-acetyl-β-d-mannosaminuronic acid, that carries a C-4-O-acetyl, an N-acetyl-α-d-fucosamine, and an N-acetyl-α-l-fucosamine. Synthetic oligosaccharides are valuable tools to unravel the immunogenicity of bacterial oligosaccharides at the molecular level. However, the rare monosaccharides, cis-glycosidic linkages, and O-acetylation represent significant challenges for the synthesis of CP8 fragments. Here the stereoselective assembly of well-defined CP8 fragments, comprising a trimer, hexamer, nonamer, and dodecamer, is presented. This is the first time that fragments larger than a single repeating trisaccharide, which has been proven to be insufficient for antigenic activity, have been assembled. Structural studies have revealed a linear conformation for the oligosaccharides, with each trisaccharide repeat tilted ∼90° with respect to the flanking repeats, which is stabilized by the acetyl groups that prevent rotation around the glycosidic linkages. The N-acetyl groups in each repeating unit point in the same direction, generating a hydrophobic flank in the trisaccharide repeats. We applied the oligomers to generate model glycoconjugate vaccine modalities, which we then used to raise anti-CP8 antibodies. The antibody interaction and immunization studies have revealed a clear length dependent structure-activity relationship for the oligosaccharides, with an oligosaccharide of at least three repeating units required for an adequate immune response.
Collapse
Affiliation(s)
- Kitt Emilie Østerlid
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Charlotte Sorieul
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Luca Unione
- Center
for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research
and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain
- Ikerbasque,
Basque Foundation for Science, Bilbao 48009, Spain
| | - Sizhe Li
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Cristian García-Sepúlveda
- Center
for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research
and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain
| | | | | | - Francesca Berni
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | - Ana Arda
- Center
for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research
and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain
- Ikerbasque,
Basque Foundation for Science, Bilbao 48009, Spain
| | - Herman S. Overkleeft
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| | | | | | - Jesús Jiménez-Barbero
- Center
for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research
and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain
- Ikerbasque,
Basque Foundation for Science, Bilbao 48009, Spain
- Department
of Organic & Inorganic Chemistry, Faculty of Science and Technology, University of the Basque Country, EHU-UPV, 48940 Leioa, Bizkaia, Spain
- Centro
de
Investigacion Biomedica En Red de Enfermedades Respiratorias, 28029 Madrid, Spain
| | | | - Jeroen D. C. Codée
- Leiden
Institute of Chemistry, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
| |
Collapse
|
2
|
Stanisic T, Ewing EU, Lindell A, Al-Jaberi F, Kongsbak-Wismann M. Vitamin D 3-VDR and vitamin A-RAR affect IL-13 and IFNγ secretion from human CD4 + T cells directly and indirectly via competition for their shared co-receptor RXR. Scand J Immunol 2025; 101:e13429. [PMID: 39822032 DOI: 10.1111/sji.13429] [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: 08/27/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 01/19/2025]
Abstract
The effects of vitamin D and vitamin A in immune cells are mediated through the vitamin D receptor (VDR) and retinoic acid receptor (RAR), respectively. These receptors share the retinoid X receptor (RXR) co-factor for transcriptional regulation. We investigated the effects of active vitamin D3 (1,25(OH)2D3) and 9-cis retinoic acid (9cRA) on T helper (TH)1 and TH2 cytokines and transcription factors in primary human blood-derived CD4+ T cells. We aimed to address the discrepancies in this field, particularly regarding the effects of 9cRA and the vitamins in combination. 1,25(OH)2D3 upregulated IL-13 and suppressed IFNγ, while 9cRA had the opposite effects. This was largely independent of a TH1/TH2 phenotype shift. Combined vitamin supplementation produced intermediate cytokine levels, not only through transcriptional regulation by VDR-RXR and RAR-RXR but also through 1,25(OH)2D3 counteracting the effects of 9cRA on solely 9cRA-responsive genes. Similar results were observed in hereditary vitamin D-resistant rickets (HVDRR) patient T cells, where VDR cannot bind to DNA, indicating that RXR binding to either receptor can limit the other's activity. Additionally, we observed downregulated RAR upon 9cRA supplementation and its re-localization out of the nucleus upon 1,25(OH)2D3 supplementation, suggesting a mechanism of indirect regulation by VDR. VDR protein levels were also upregulated upon 9cRA supplementation, suggesting a novel negative feedback mechanism of 9cRA transcriptional activity, whereby 9cRA promotes its own competitor. This study sets the stage for future research into the combined immunomodulatory mechanisms of 1,25(OH)2D3 and 9cRA, involving both direct transcriptional regulation and indirect regulation via RXR competitive binding.
Collapse
Affiliation(s)
- Tiana Stanisic
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emma Uttrup Ewing
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alma Lindell
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Fatima Al-Jaberi
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Martin Kongsbak-Wismann
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
3
|
McReynolds AKG, Pagella EA, Ridder MJ, Rippee O, Clark Z, Rekowski MJ, Pritchard MT, Bose JL. YjbH contributes to Staphylococcus aureus skin pathology and immune response through Agr-mediated α-toxin regulation. Virulence 2024; 15:2399798. [PMID: 39229975 PMCID: PMC11404607 DOI: 10.1080/21505594.2024.2399798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 09/05/2024] Open
Abstract
Staphylococcus aureus is the most common cause of skin and soft tissue infections (SSTIs) with Methicillin-Resistant S. aureus (MRSA) strains being a major contributor in both community and hospital settings. S. aureus relies on metabolic diversity and a large repertoire of virulence factors to cause disease. This includes α-hemolysin (Hla), an integral player in tissue damage found in various models, including SSTIs. Previously, we identified a role for the Spx adapter protein, YjbH, in the regulation of several virulence factors and as an inhibitor of pathogenesis in a sepsis model. In this study, we found that YjbH is critical for tissue damage during SSTI, and its absence leads to decreased proinflammatory chemokines and cytokines in the skin. We identified no contribution of YjbI, encoded on the same transcript as YjbH. Using a combination of reporters and quantitative hemolysis assays, we demonstrated that YjbH impacts Hla expression and activity both in vitro and in vivo. Additionally, expression of Hla from a non-native promoter reversed the tissue damage phenotype of the ΔyjbIH mutant. Lastly, we identified reduced Agr activity as the likely cause for reduced Hla production in the ΔyjbH mutant. This work continues to define the importance of YjbH in the pathogenesis of S. aureus infection as well as identify a new pathway important for Hla production.
Collapse
Affiliation(s)
- Aubrey K. G. McReynolds
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Emma A. Pagella
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Miranda J. Ridder
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Olivia Rippee
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Zachary Clark
- The Mass Spectrometry and Proteomics Core, University of Kansas Medical Center, Kansas City, KS, USA
| | - Michaella J. Rekowski
- The Mass Spectrometry and Proteomics Core, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Michele T. Pritchard
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jeffrey L. Bose
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
4
|
Yan G, Liu Z, Teng T, Dong W, Lan T, Fan J, Tang K, Qin S, Nie W. Metagenomic next-generation sequencing of osteoarticular tissue for the diagnosis of suspected osteoarticular tuberculosis. Microbiol Spectr 2024; 12:e0359823. [PMID: 39513695 PMCID: PMC11619600 DOI: 10.1128/spectrum.03598-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 10/02/2024] [Indexed: 11/15/2024] Open
Abstract
The objective of the study is to determine the accuracy of metagenomic next-generation sequencing (mNGS) in diagnosing osteoarticular tuberculosis (TB) infection and to compare it with mycobacteria growth indicator tube (MGIT) and Xpert assays. We retrospectively analyzed 162 patients admitted with suspected osteoarticular TB. Osteoarticular tissue (66.67%) and abscess specimens (33.33%) from patients were tested for MGIT, GeneXpert/RIF, and mNGS. mNGS assay detected 76 cases (46.9%) with bacterial, 63 cases (38.9%) with mycobacterial, 22 cases (13.6%) with fungal, and 1 case (0.6%) with actinomycetal organisms. These 162 pathogens were classified into 21 species. The most frequent species detected was Mycobacterium tuberculosis complex (29.0%), followed by Staphylococcus aureus (20.4%), Mycobacterium abscessus (5.6%), and Candida albicans (5.6%). Taking the "gold standard" TB diagnosis as the standard, the positive predictive values of mNGS, Xpert, and MGIT culture were both 100.00%. The negative predictive values of mNGS, Xpert, and MGIT culture and assays were 94.26%, 98.29%, and 88.46%, respectively. The sensitivity of mNGS detection (85.11%) was similar to that of Xpert (95.74%) and higher than that of MGIT culture (68.08%). The specificities of mNGS detection, Xpert, and MGIT culture were both 100.00%. The area under the curve value of the mNGS assay was 0.895 (95% CI: 0.830, 0.960), which was greater than that of the MGIT culture-based assay of 0.840 (95% CI: 0.757, 0.924), which was similar to 0.979 (95% CI: 0.945, 1.000) for Xpert assay. The pathogen detection rate of mNGS in diagnosing suspected osteoarticular TB exceeded that of conventional methods. IMPORTANCE In the detection of unknown infectious disease pathogens, the overall efficacy of traditional detection methods, such as culture, is low, and traditional PCR testing is also limited to the gene sequences of known pathogenic microorganisms. Metagenomic next-generation sequencing (mNGS) performs DNA sequencing by studying the entire microbial community genome in a given sample, without the need for isolation and culture. Previous studies have shown that mNGS performs better on pulmonary and extrapulmonary samples when compared with Xpert, traditional pathogenetic tests, and even parallel diagnostics. However, it should be emphasized that only a few studies have explored the performance of mNGS in detecting Mycobacterium tuberculosis in clinical samples associated with bone and joint infections. We conducted this retrospective study to provide additional data to support the use of mNGS in the clinical setting to identify pathogens within abscesses or tissue samples associated with bone and joint infections.
Collapse
Affiliation(s)
- Guangxuan Yan
- Orthopedics Department, Beijing Chest Hospital affiliated to Capital Medical University, Beijing Tuberculosis & Thoracic Tumor Research Institute, Beijing, China
| | - Zhifeng Liu
- Beijing Emercency Mecial Center, Beijing, China
| | - Tianlu Teng
- Respiratory Department, Beijing Chest Hospital affiliated to Capital Medical University, Beijing, China
| | - Weijie Dong
- Orthopedics Department, Beijing Chest Hospital affiliated to Capital Medical University, Beijing Tuberculosis & Thoracic Tumor Research Institute, Beijing, China
| | - Tinglong Lan
- Orthopedics Department, Beijing Chest Hospital affiliated to Capital Medical University, Beijing Tuberculosis & Thoracic Tumor Research Institute, Beijing, China
| | - Jun Fan
- Orthopedics Department, Beijing Chest Hospital affiliated to Capital Medical University, Beijing Tuberculosis & Thoracic Tumor Research Institute, Beijing, China
| | - Kai Tang
- Orthopedics Department, Beijing Chest Hospital affiliated to Capital Medical University, Beijing Tuberculosis & Thoracic Tumor Research Institute, Beijing, China
| | - Shibing Qin
- Orthopedics Department, Beijing Chest Hospital affiliated to Capital Medical University, Beijing Tuberculosis & Thoracic Tumor Research Institute, Beijing, China
| | - Wenjuan Nie
- Tuberculosis Department, Beijing Chest Hospital affiliated to Capital Medical University, Beijing Tuberculosis & Thoracic Tumor Research Institute, Beijing, China
| |
Collapse
|
5
|
Bell RD, Cann EA, Mishra B, Valencia M, Zhang Q, Huang M, Yang X, Carli A, Bostrom M, Ivashkiv LB. Staphyloccocus aureus biofilm, in absence of planktonic bacteria, produces factors that activate counterbalancing inflammatory and immune-suppressive genes in human monocytes. J Orthop Res 2024; 42:2582-2592. [PMID: 38922976 PMCID: PMC11481048 DOI: 10.1002/jor.25919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 04/25/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024]
Abstract
Staphyloccocus aureus (S. aureus) is a major bacterial pathogen in orthopedic periprosthetic joint infection (PJI). S. aureus forms biofilms that promote persistent infection by shielding bacteria from immune cells and inducing an antibiotic-tolerant metabolic state. We developed an in vitro system to study S. aureus biofilm interactions with primary human monocytes in the absence of planktonic bacteria. In line with previous in vivo data, S. aureus biofilm induced expression of inflammatory genes such as TNF and IL1B, and their anti-inflammatory counter-regulator IL10. S. aureus biofilm also activated expression of PD-1 ligands, and IL-1RA, molecules that have the potential to suppress T cell function or differentiation of protective Th17 cells. Gene induction did not require monocyte:biofilm contact and was mediated by a soluble factor(s) produced by biofilm-encased bacteria that was heat resistant and >3 kD in size. Activation of suppressive genes by biofilm was sensitive to suppression by Jak kinase inhibition. These results support an evolving paradigm that biofilm plays an active role in modulating immune responses, and suggest this occurs via production of a soluble vita-pathogen-associated molecular pattern, a molecule that signals microbial viability. Induction of T cell suppressive genes by S. aureus biofilm provides insights into mechanisms that can suppress T cell immunity in PJI.
Collapse
Affiliation(s)
- Richard D Bell
- Research Institute and Arthritis and Tissue Degeneration Program, Hospital for Special Surgery
| | - E. Abrefi Cann
- Research Institute and Arthritis and Tissue Degeneration Program, Hospital for Special Surgery
| | - Bikash Mishra
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine
| | - Melanie Valencia
- Research Institute and Arthritis and Tissue Degeneration Program, Hospital for Special Surgery
| | - Qiong Zhang
- Research Institute and Arthritis and Tissue Degeneration Program, Hospital for Special Surgery
| | - Mary Huang
- Research Institute and Arthritis and Tissue Degeneration Program, Hospital for Special Surgery
| | - Xu Yang
- Research Institute and Arthritis and Tissue Degeneration Program, Hospital for Special Surgery
| | - Alberto Carli
- Department of Orthopedic Surgery, Hospital for Special Surgery
| | - Mathias Bostrom
- Department of Orthopedic Surgery, Hospital for Special Surgery
| | - Lionel B Ivashkiv
- Research Institute and Arthritis and Tissue Degeneration Program, Hospital for Special Surgery
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine
| |
Collapse
|
6
|
Appel K, Rose T, Zimmermann C, Günnewich N. In Vitro Anti-inflammatory Effects of Larch Turpentine, Turpentine Oil, Eucalyptus Oil, and Their Mixture as Contained in a Marketed Ointment. PLANTA MEDICA 2024; 90:1023-1029. [PMID: 39260387 PMCID: PMC11614573 DOI: 10.1055/a-2388-7527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 08/01/2024] [Indexed: 09/13/2024]
Abstract
An ointment containing larch turpentine, turpentine oil, and eucalyptus oil has been used for almost a century for the symptomatic treatment of mild, localized, purulent inflammations of the skin. Its clinical efficacy in the treatment of skin infections has been shown in clinical trials, but the mode of action of the active ingredients on inflammation is not known. We studied the anti-inflammatory properties of the active ingredients of the ointment and their mixture in a human monocyte cell model, in which the cells were stimulated with lipopolysaccharide and incubated with the test substances. The cytotoxic threshold of each test substance and the mixture was identified using the alamarBlue assay, and their anti-inflammatory activity was assessed by measuring the release of interleukins IL-1β, IL-6, IL-8, monocyte chemoattractant protein-1, prostaglandin E2, and TNF-α. Cell toxicity was observed at a mixture concentration of 10 µg/mL. All immunological assays were carried out at nontoxic concentrations. Larch turpentine decreased IL-1β, monocyte chemoattractant protein-1, and prostaglandin E2 release at a concentration of 3.9 µg/mL and TNF-α at concentrations > 1.95 µg/mL, whereas eucalyptus oil and turpentine oil had no relevant inhibitory effects. The mixture dose-dependently inhibited IL-1β, IL-6, monocyte chemoattractant protein-1, prostaglandin E2, and TNF-α release at concentrations > 1 µg/mL. IL-8 release was only marginally affected. The anti-inflammatory activity of the herbal ingredients and their mixture was confirmed in this model. This effect seems to be mediated mainly by larch turpentine, with turpentine oil and eucalyptus oil exerting an additive or possibly synergistic function.
Collapse
Affiliation(s)
- Kurt Appel
- VivaCell Biotechnology GmbH, Denzlingen, Germany
| | | | | | | |
Collapse
|
7
|
Subbarayudu S, Namasivayam SKR, Arockiaraj J. Immunomodulation in Non-traditional Therapies for Methicillin-resistant Staphylococcus aureus (MRSA) Management. Curr Microbiol 2024; 81:346. [PMID: 39240286 DOI: 10.1007/s00284-024-03875-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
The rise of methicillin-resistant Staphylococcus aureus (MRSA) poses a significant challenge in clinical settings due to its ability to evade conventional antibiotic treatments. This overview explores the potential of immunomodulatory strategies as alternative therapeutic approaches to combat MRSA infections. Traditional antibiotics are becoming less effective, necessitating innovative solutions that harness the body's immune system to enhance pathogen clearance. Recent advancements in immunotherapy, including the use of antimicrobial peptides, phage therapy, and mechanisms of immune cells, demonstrate promise in enhancing the body's ability to clear MRSA infections. However, the exact interactions between these therapies and immunomodulation are not fully understood, underscoring the need for further research. Hence, this review aims to provide a broad overview of the current understanding of non-traditional therapeutics and their impact on immune responses, which could lead to more effective MRSA treatment strategies. Additionally, combining immunomodulatory agents with existing antibiotics may improve outcomes, particularly for immunocompromised patients or those with chronic infections. As the landscape of antibiotic resistance evolves, the development of effective immunotherapeutic strategies could play a vital role in managing MRSA infections and reducing reliance on traditional antibiotics. Future research must focus on optimizing these approaches and validating their efficacy in diverse clinical populations to address the urgent need for effective MRSA management strategies.
Collapse
Affiliation(s)
- Suthi Subbarayudu
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, 603203, India
| | - S Karthick Raja Namasivayam
- Centre for Applied Research, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu, 602105, India.
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu, 603203, India.
| |
Collapse
|
8
|
Reynolds MB, Klein B, McFadden MJ, Judge NK, Navarrete HE, Michmerhuizen BC, Awad D, Schultz TL, Harms PW, Zhang L, O'Meara TR, Sexton JZ, Lyssiotis CA, Kahlenberg JM, O'Riordan MX. Type I interferon governs immunometabolic checkpoints that coordinate inflammation during Staphylococcal infection. Cell Rep 2024; 43:114607. [PMID: 39126652 PMCID: PMC11590196 DOI: 10.1016/j.celrep.2024.114607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/09/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Macrophage metabolic plasticity is central to inflammatory programming, yet mechanisms of coordinating metabolic and inflammatory programs during infection are poorly defined. Here, we show that type I interferon (IFN) temporally guides metabolic control of inflammation during methicillin-resistant Staphylococcus aureus (MRSA) infection. We find that staggered Toll-like receptor and type I IFN signaling in macrophages permit a transient energetic state of combined oxidative phosphorylation (OXPHOS) and aerobic glycolysis followed by inducible nitric oxide synthase (iNOS)-mediated OXPHOS disruption. This disruption promotes type I IFN, suppressing other pro-inflammatory cytokines, notably interleukin-1β. Upon infection, iNOS expression peaks at 24 h, followed by lactate-driven Nos2 repression via histone lactylation. Type I IFN pre-conditioning prolongs infection-induced iNOS expression, amplifying type I IFN. Cutaneous MRSA infection in mice constitutively expressing epidermal type I IFN results in elevated iNOS levels, impaired wound healing, vasculopathy, and lung infection. Thus, kinetically regulated type I IFN signaling coordinates immunometabolic checkpoints that control infection-induced inflammation.
Collapse
Affiliation(s)
- Mack B Reynolds
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Benjamin Klein
- Department of Internal Medicine, Division of Rheumatology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Michael J McFadden
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Norah K Judge
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Hannah E Navarrete
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Britton C Michmerhuizen
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Dominik Awad
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Tracey L Schultz
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Paul W Harms
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Li Zhang
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Teresa R O'Meara
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jonathan Z Sexton
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - J Michelle Kahlenberg
- Department of Internal Medicine, Division of Rheumatology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Mary X O'Riordan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
9
|
Lang JC, Brutscher A, Ehrström M, Melican K. Tissue resident cells differentiate S. aureus from S. epidermidis via IL-1β following barrier disruption in healthy human skin. PLoS Pathog 2024; 20:e1012056. [PMID: 39208402 PMCID: PMC11389914 DOI: 10.1371/journal.ppat.1012056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 09/11/2024] [Accepted: 08/11/2024] [Indexed: 09/04/2024] Open
Abstract
The Staphylococcus sp. are a dominant part of the human skin microbiome and present across the body. Staphylococcus epidermidis is a ubiquitous skin commensal, while S. aureus is thought to colonize at least 30% of the population. S. aureus are not only colonizers but a leading cause of skin and soft tissue infections and a critical healthcare concern. To understand how healthy human skin may differentiate commensal bacteria, such as S. epidermidis, from the potential pathogen methicillin-resistant S. aureus (MRSA), we use ex vivo human skin models that allow us to study this host-bacterial interaction in the most clinically relevant environment. Our work highlights the role of the outer stratum corneum as a protective physical barrier against invasion by colonizing Staphylococci. We show how the structural cells of the skin can internalize and respond to different Staphylococci with increasing sensitivity. In intact human skin, a discriminatory IL-1β response was identified, while disruption of the protective stratum corneum triggered an increased and more diverse immune response. We identified and localized tissue resident Langerhans cells (LCs) as a potential source of IL-1β and go on to show a dose-dependent response of MUTZ-LCs to S. aureus but not S. epidermidis. This suggests an important role of LCs in sensing and discriminating between bacteria in healthy human skin, particularly in intact skin and provides a detailed snapshot of how human skin differentiates between friend and potential foe. With the rise in antibiotic resistance, understanding the innate immune response of healthy skin may help us find ways to enhance or manipulate these natural defenses to prevent invasive infection.
Collapse
Affiliation(s)
- Julia C Lang
- AIMES-Center for the Advancement of Integrated Medical and Engineering Sciences, Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Andreas Brutscher
- AIMES-Center for the Advancement of Integrated Medical and Engineering Sciences, Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Keira Melican
- AIMES-Center for the Advancement of Integrated Medical and Engineering Sciences, Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
Sozzani S, Sozio F, Del Prete A. Chemerin is a key player in antimicrobial defense in skin. Cell Mol Immunol 2024; 21:638-640. [PMID: 38755456 PMCID: PMC11143294 DOI: 10.1038/s41423-024-01159-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 05/18/2024] Open
Affiliation(s)
- Silvano Sozzani
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Institute Pasteur-Italia, Rome, Italy.
| | - Francesca Sozio
- Department of Surgery, Sapienza University of Rome, Rome, Italy
| | - Annalisa Del Prete
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
11
|
Chen Y, Song Y, Wang Z, Lai Y, Yin W, Cai Q, Han M, Cai Y, Xue Y, Chen Z, Li X, Chen J, Li M, Li H, He R. The chemerin-CMKLR1 axis in keratinocytes impairs innate host defense against cutaneous Staphylococcus aureus infection. Cell Mol Immunol 2024; 21:533-545. [PMID: 38532043 PMCID: PMC11143357 DOI: 10.1038/s41423-024-01152-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 03/01/2024] [Indexed: 03/28/2024] Open
Abstract
The skin is the most common site of Staphylococcus aureus infection, which can lead to various diseases, including invasive and life-threatening infections, through evasion of host defense. However, little is known about the host factors that facilitate the innate immune evasion of S. aureus in the skin. Chemerin, which is abundantly expressed in the skin and can be activated by proteases derived from S. aureus, has both direct bacteria-killing activity and immunomodulatory effects via interactions with its receptor CMKLR1. Here, we demonstrate that a lack of the chemerin/CMKLR1 axis increases the neutrophil-mediated host defense against S. aureus in a mouse model of cutaneous infection, whereas chemerin overexpression, which mimics high levels of chemerin in obese individuals, exacerbates S. aureus cutaneous infection. Mechanistically, we identified keratinocytes that express CMKLR1 as the main target of chemerin to suppress S. aureus-induced IL-33 expression, leading to impaired skin neutrophilia and bacterial clearance. CMKLR1 signaling specifically inhibits IL-33 expression induced by cell wall components but not secreted proteins of S. aureus by inhibiting Akt activation in mouse keratinocytes. Thus, our study revealed that the immunomodulatory effect of the chemerin/CMKLR1 axis mediates innate immune evasion of S. aureus in vivo and likely increases susceptibility to S. aureus infection in obese individuals.
Collapse
Affiliation(s)
- Yu Chen
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE/NHC), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yan Song
- Department of Laboratory Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Zhe Wang
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE/NHC), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Yangfan Lai
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE/NHC), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Wei Yin
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE/NHC), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Qian Cai
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE/NHC), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Miaomiao Han
- Allergy Center, Department of Otolaryngology, Affiliated Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Yiheng Cai
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE/NHC), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Yushan Xue
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE/NHC), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China
| | - Zhengrong Chen
- Department of Respiratory Diseases, Children's Hospital of Soochow University, Suzhou, China
| | - Xi Li
- Biology Science Institutes, Chongqing Medical University, Chongqing, 400032, China
| | - Jing Chen
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Department of Nephrology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Min Li
- Department of Laboratory Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Faculty of Medical Laboratory Science, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Huabin Li
- Allergy Center, Department of Otolaryngology, Affiliated Eye and ENT Hospital, Fudan University, Shanghai, 200031, China.
| | - Rui He
- Department of Immunology, Key Laboratory of Medical Molecular Virology (MOE/NHC), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, 200032, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China.
- Research Center of Allergy and Diseases, Fudan University, 200040, Shanghai, China.
| |
Collapse
|
12
|
Hajam IA, Liu GY. Linking S. aureus Immune Evasion Mechanisms to Staphylococcal Vaccine Failures. Antibiotics (Basel) 2024; 13:410. [PMID: 38786139 PMCID: PMC11117348 DOI: 10.3390/antibiotics13050410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024] Open
Abstract
Vaccination arguably remains the only long-term strategy to limit the spread of S. aureus infections and its related antibiotic resistance. To date, however, all staphylococcal vaccines tested in clinical trials have failed. In this review, we propose that the failure of S. aureus vaccines is intricately linked to prior host exposure to S. aureus and the pathogen's capacity to evade adaptive immune defenses. We suggest that non-protective immune imprints created by previous exposure to S. aureus are preferentially recalled by SA vaccines, and IL-10 induced by S. aureus plays a unique role in shaping these non-protective anti-staphylococcal immune responses. We discuss how S. aureus modifies the host immune landscape, which thereby necessitates alternative approaches to develop successful staphylococcal vaccines.
Collapse
Affiliation(s)
- Irshad Ahmed Hajam
- Department of Pediatrics, University of California San Diego, San Diego, CA 92093, USA;
| | - George Y. Liu
- Department of Pediatrics, University of California San Diego, San Diego, CA 92093, USA;
- Division of Infectious Diseases, Rady Children’s Hospital, San Diego, CA 92123, USA
| |
Collapse
|
13
|
Kurz H, Lehmberg K, Farmand S. Inborn errors of immunity with susceptibility to S. aureus infections. Front Pediatr 2024; 12:1389650. [PMID: 38720948 PMCID: PMC11078099 DOI: 10.3389/fped.2024.1389650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 03/25/2024] [Indexed: 05/12/2024] Open
Abstract
Staphylococcus aureus (S. aureus) is a significant human pathogen, in particular in patients with an underlying medical condition. It is equipped with a large variety of virulence factors enabling both colonization and invasive disease. The spectrum of manifestation is broad, ranging from superficial skin infections to life-threatening conditions like pneumonia and sepsis. As a major cause of healthcare-associated infections, there is a great need in understanding staphylococcal immunity and defense mechanisms. Patients with inborn errors of immunity (IEI) frequently present with pathological infection susceptibility, however, not all of them are prone to S. aureus infection. Thus, enhanced frequency or severity of S. aureus infections can serve as a clinical indicator of a specific underlying immunological impairment. In addition, the analysis of immunological functions in patients with susceptibility to S. aureus provides a unique opportunity of understanding the complex interplay between staphylococcal virulence and host immune predisposition. While the importance of quantitatively and qualitatively normal neutrophils is widely known, less awareness exists about the role of specific cytokines such as functional interleukin (IL)-6 signaling. This review categorizes well-known IEI in light of their susceptibility to S. aureus and discusses the relevant associated pathomechanisms. Understanding host-pathogen-interactions in S. aureus infections in susceptible individuals can pave the way for more effective management and preventive treatment options. Moreover, these insights might help to identify patients who should be screened for an underlying IEI. Ultimately, enhanced understanding of pathogenesis and immune responses in S. aureus infections may also be of relevance for the general population.
Collapse
Affiliation(s)
- Hannah Kurz
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kai Lehmberg
- Division of Pediatric Stem Cell Transplantation and Immunology, Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Susan Farmand
- Division of Pediatric Stem Cell Transplantation and Immunology, Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
14
|
Dean CJ, Peña-Mosca F, Ray T, Wehri TJ, Sharpe K, Antunes, Jr. AM, Doster E, Fernandes L, Calles VF, Bauman C, Godden S, Heins B, Pinedo P, Machado VS, Caixeta LS, Noyes NR. Exploring associations between the teat apex metagenome and Staphylococcus aureus intramammary infections in primiparous cows under organic directives. Appl Environ Microbiol 2024; 90:e0223423. [PMID: 38497641 PMCID: PMC11022539 DOI: 10.1128/aem.02234-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/04/2024] [Indexed: 03/19/2024] Open
Abstract
The primary objective of this study was to identify associations between the prepartum teat apex microbiome and the presence of Staphylococcus aureus intramammary infections (IMI) in primiparous cows during the first 5 weeks after calving. We performed a case-control study using shotgun metagenomics of the teat apex and culture-based milk data collected longitudinally from 710 primiparous cows on five organic dairy farms. Cases had higher odds of having S. aureus metagenomic DNA on the teat apex prior to parturition compared to controls (OR = 38.9, 95% CI: 14.84-102.21). Differential abundance analysis confirmed this association, with cases having a 23.8 higher log fold change (LFC) in the abundance of S. aureus in their samples compared to controls. Of the most prevalent microorganisms in controls, those associated with a lower risk of post-calving S. aureus IMI included Microbacterium phage Min 1 (OR = 0.37, 95% CI: 0.25-0.53), Corynebacterium efficiens (OR = 0.53, 95% CI: 0.30-0.94), Kocuria polaris (OR = 0.54, 95% CI: 0.35-0.82), Micrococcus terreus (OR = 0.64, 95% CI: 0.44-0.93), and Dietzia alimentaria (OR = 0.45, 95% CI: 0.26-0.75). Genes encoding for Microcin B17 AMPs were the most prevalent on the teat apex of cases and controls (99.7% in both groups). The predicted abundance of genes encoding for Microcin B17 was also higher in cases compared to controls (LFC 0.26). IMPORTANCE Intramammary infections (IMI) caused by Staphylococcus aureus remain an important problem for the dairy industry. The microbiome on the external skin of the teat apex may play a role in mitigating S. aureus IMI risk, in particular the production of antimicrobial peptides (AMPs) by commensal microbes. However, current studies of the teat apex microbiome utilize a 16S approach, which precludes the detection of genomic features such as genes that encode for AMPs. Therefore, further research using a shotgun metagenomic approach is needed to understand what role prepartum teat apex microbiome dynamics play in IMI risk.
Collapse
Affiliation(s)
- C. J. Dean
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - F. Peña-Mosca
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - T. Ray
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - T. J. Wehri
- Department of Animal Science, University of Minnesota, St. Paul, Minnesota, USA
| | - K. Sharpe
- Department of Animal Science, University of Minnesota, St. Paul, Minnesota, USA
| | - A. M. Antunes, Jr.
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - E. Doster
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - L. Fernandes
- Department of Veterinary Sciences, Texas Tech University, Lubbock, Texas, USA
| | - V. F. Calles
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - C. Bauman
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - S. Godden
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - B. Heins
- Department of Animal Science, University of Minnesota, St. Paul, Minnesota, USA
| | - P. Pinedo
- Department of Animal Science, Colorado State University, Fort Collins, Colorado, USA
| | - V. S. Machado
- Department of Veterinary Sciences, Texas Tech University, Lubbock, Texas, USA
| | - L. S. Caixeta
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - N. R. Noyes
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, Minnesota, USA
| |
Collapse
|
15
|
Clowry J, Dempsey DJ, Claxton TJ, Towell AM, Turley MB, Sutton M, Geoghegan JA, Kezic S, Jakasa I, White A, Irvine AD, McLoughlin RM. Distinct T cell signatures are associated with Staphylococcus aureus skin infection in pediatric atopic dermatitis. JCI Insight 2024; 9:e178789. [PMID: 38716729 PMCID: PMC11141913 DOI: 10.1172/jci.insight.178789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 04/03/2024] [Indexed: 06/02/2024] Open
Abstract
Atopic dermatitis (AD) is an inflammatory skin condition with a childhood prevalence of up to 25%. Microbial dysbiosis is characteristic of AD, with Staphylococcus aureus the most frequent pathogen associated with disease flares and increasingly implicated in disease pathogenesis. Therapeutics to mitigate the effects of S. aureus have had limited efficacy and S. aureus-associated temporal disease flares are synonymous with AD. An alternative approach is an anti-S. aureus vaccine, tailored to AD. Experimental vaccines have highlighted the importance of T cells in conferring protective anti-S. aureus responses; however, correlates of T cell immunity against S. aureus in AD have not been identified. We identify a systemic and cutaneous immunological signature associated with S. aureus skin infection (ADS.aureus) in a pediatric AD cohort, using a combined Bayesian multinomial analysis. ADS.aureus was most highly associated with elevated cutaneous chemokines IP10 and TARC, which preferentially direct Th1 and Th2 cells to skin. Systemic CD4+ and CD8+ T cells, except for Th2 cells, were suppressed in ADS.aureus, particularly circulating Th1, memory IL-10+ T cells, and skin-homing memory Th17 cells. Systemic γδ T cell expansion in ADS.aureus was also observed. This study suggests that augmentation of protective T cell subsets is a potential therapeutic strategy in the management of S. aureus in AD.
Collapse
Affiliation(s)
- Julianne Clowry
- Department of Dermatology, National Children’s Research Centre, Children’s Health Ireland at Crumlin, Dublin, Ireland
- Clinical Medicine, Trinity College Dublin, Dublin, Ireland
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Daniel J. Dempsey
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Tracey J. Claxton
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Aisling M. Towell
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Mary B. Turley
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Martin Sutton
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Joan A. Geoghegan
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Sanja Kezic
- Amsterdam UMC, University of Amsterdam, Department of Public and Occupational Health, Amsterdam Public Health Research Institute, Amsterdam, Netherlands
| | - Ivone Jakasa
- Laboratory for Analytical Chemistry, Department of Chemistry and Biochemistry, Faculty of Food Technology and Biotechnology, University of Zagreb, Zagreb, Croatia
| | - Arthur White
- School of Computer Science and Statistics, Trinity College Dublin, Dublin, Ireland
| | - Alan D. Irvine
- Department of Dermatology, National Children’s Research Centre, Children’s Health Ireland at Crumlin, Dublin, Ireland
- Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| | - Rachel M. McLoughlin
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
16
|
Cavagnero KJ, Li F, Dokoshi T, Nakatsuji T, O’Neill AM, Aguilera C, Liu E, Shia M, Osuoji O, Hata T, Gallo RL. CXCL12+ dermal fibroblasts promote neutrophil recruitment and host defense by recognition of IL-17. J Exp Med 2024; 221:e20231425. [PMID: 38393304 PMCID: PMC10890925 DOI: 10.1084/jem.20231425] [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: 08/16/2023] [Revised: 11/17/2023] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
The skin provides an essential barrier for host defense through rapid action of multiple resident and recruited cell types, but the complex communication network governing these processes is incompletely understood. To define these cell-cell interactions more clearly, we performed an unbiased network analysis of mouse skin during invasive S. aureus infection and revealed a dominant role for CXCL12+ fibroblast subsets in neutrophil communication. These subsets predominantly reside in the reticular dermis, express adipocyte lineage markers, detect IL-17 and TNFα, and promote robust neutrophil recruitment through NFKBIZ-dependent release of CXCR2 ligands and CXCL12. Targeted deletion of Il17ra in mouse fibroblasts resulted in greatly reduced neutrophil recruitment and increased infection by S. aureus. Analogous human CXCL12+ fibroblast subsets abundantly express neutrophil chemotactic factors in psoriatic skin that are subsequently decreased upon therapeutic targeting of IL-17. These findings show that CXCL12+ dermal immune acting fibroblast subsets play a critical role in cutaneous neutrophil recruitment and host defense.
Collapse
Affiliation(s)
- Kellen J. Cavagnero
- Department of Dermatology, University of California, San Diego. La Jolla, CA, USA
| | - Fengwu Li
- Department of Dermatology, University of California, San Diego. La Jolla, CA, USA
| | - Tatsuya Dokoshi
- Department of Dermatology, University of California, San Diego. La Jolla, CA, USA
| | - Teruaki Nakatsuji
- Department of Dermatology, University of California, San Diego. La Jolla, CA, USA
| | - Alan M. O’Neill
- Department of Dermatology, University of California, San Diego. La Jolla, CA, USA
| | - Carlos Aguilera
- Department of Dermatology, University of California, San Diego. La Jolla, CA, USA
| | - Edward Liu
- Department of Dermatology, University of California, San Diego. La Jolla, CA, USA
| | - Michael Shia
- Department of Dermatology, University of California, San Diego. La Jolla, CA, USA
| | - Olive Osuoji
- Department of Dermatology, University of California, San Diego. La Jolla, CA, USA
| | - Tissa Hata
- Department of Dermatology, University of California, San Diego. La Jolla, CA, USA
| | - Richard L. Gallo
- Department of Dermatology, University of California, San Diego. La Jolla, CA, USA
| |
Collapse
|
17
|
Nanda N, Alphonse MP. From Host Defense to Metabolic Signatures: Unveiling the Role of γδ T Cells in Bacterial Infections. Biomolecules 2024; 14:225. [PMID: 38397462 PMCID: PMC10886488 DOI: 10.3390/biom14020225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/12/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
The growth of antibiotic-resistant bacterial infections necessitates focusing on host-derived immunotherapies. γδ T cells are an unconventional T cell subset, making up a relatively small portion of healthy circulating lymphocytes but a substantially increased proportion in mucosal and epithelial tissues. γδ T cells are activated and expanded in response to bacterial infection, having the capability to produce proinflammatory cytokines to recruit neutrophils and clear infection. They also play a significant role in dampening immune response to control inflammation and protecting the host against secondary challenge, making them promising targets when developing immunotherapy. Importantly, γδ T cells have differential metabolic states influencing their cytokine profile and subsequent inflammatory capacity. Though these differential metabolic states have not been well studied or reviewed in the context of bacterial infection, they are critical in understanding the mechanistic underpinnings of the host's innate immune response. Therefore, this review will focus on the context-specific host defense conferred by γδ T cells during infection with Staphylococcus aureus, Streptococcus pneumoniae, Listeria monocytogenes, and Mycobacterium tuberculosis.
Collapse
Affiliation(s)
| | - Martin P. Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
18
|
Zhang XE, Zheng P, Ye SZ, Ma X, Liu E, Pang YB, He QY, Zhang YX, Li WQ, Zeng JH, Guo J. Microbiome: Role in Inflammatory Skin Diseases. J Inflamm Res 2024; 17:1057-1082. [PMID: 38375021 PMCID: PMC10876011 DOI: 10.2147/jir.s441100] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/23/2024] [Indexed: 02/21/2024] Open
Abstract
As the body's largest organ, the skin harbors a highly diverse microbiota, playing a crucial role in resisting foreign pathogens, nurturing the immune system, and metabolizing natural products. The dysregulation of human skin microbiota is implicated in immune dysregulation and inflammatory responses. This review delineates the microbial alterations and immune dysregulation features in common Inflammatory Skin Diseases (ISDs) such as psoriasis, rosacea, atopic dermatitis(AD), seborrheic dermatitis(SD), diaper dermatitis(DD), and Malassezia folliculitis(MF).The skin microbiota, a complex and evolving community, undergoes changes in composition and function that can compromise the skin microbial barrier. These alterations induce water loss and abnormal lipid metabolism, contributing to the onset of ISDs. Additionally, microorganisms release toxins, like Staphylococcus aureus secreted α toxins and proteases, which may dissolve the stratum corneum, impairing skin barrier function and allowing entry into the bloodstream. Microbes entering the bloodstream activate molecular signals, leading to immune disorders and subsequent skin inflammatory responses. For instance, Malassezia stimulates dendritic cells(DCs) to release IL-12 and IL-23, differentiating into a Th17 cell population and producing proinflammatory mediators such as IL-17, IL-22, TNF-α, and IFN-α.This review offers new insights into the role of the human skin microbiota in ISDs, paving the way for future skin microbiome-specific targeted therapies.
Collapse
Affiliation(s)
- Xue-Er Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 6610075, People’s Republic of China
| | - Pai Zheng
- Chengdu University of Traditional Chinese Medicine, Chengdu, 6610075, People’s Republic of China
| | - Sheng-Zhen Ye
- Chengdu University of Traditional Chinese Medicine, Chengdu, 6610075, People’s Republic of China
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 6610072, People’s Republic of China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People’s Republic of China
| | - E Liu
- Chengdu University of Traditional Chinese Medicine, Chengdu, 6610075, People’s Republic of China
| | - Yao-Bin Pang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 6610075, People’s Republic of China
| | - Qing-Ying He
- Chengdu University of Traditional Chinese Medicine, Chengdu, 6610075, People’s Republic of China
| | - Yu-Xiao Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 6610075, People’s Republic of China
| | - Wen-Quan Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, 6610075, People’s Republic of China
| | - Jin-Hao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, People’s Republic of China
| | - Jing Guo
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 6610072, People’s Republic of China
| |
Collapse
|
19
|
Vasudevan S, David H, Chanemougam L, Ramani J, Ramesh Sangeetha M, Solomon AP. Emergence of persister cells in Staphylococcus aureus: calculated or fortuitous move? Crit Rev Microbiol 2024; 50:64-75. [PMID: 36548910 DOI: 10.1080/1040841x.2022.2159319] [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: 11/02/2021] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
A stable but reversible phenotype switch from normal to persister state is advantageous to the intracellular pathogens to cause recurrent infections and to evade the host immune system. Staphylococcus aureus is a versatile opportunistic pathogen known to cause chronic infections with significant mortality. One of the notable features is the ability to switch to a per-sisters cell, which is found in planktonic and biofilm states. This phenotypic switch is always an open question to explore the hidden fundamental science that coheres with a calculated or fortuitous move. Toxin-antitoxin modules, nutrient stress, and an erroneous translation-enabled state of dormancy entail this persistent behaviour in S. aureus. It is paramount to get a clear picture of why the cell chooses to enter a persistent condition, as it would decide the course of treatment. Analyzing the exit from a persistent state to an active state and the subsequent repercussion of this transition is essential to determine its role in chronic infections. This review attempts to provide a constructed argument discussing the most widely accepted mechanisms and identifying the various attributes of persistence.
Collapse
Affiliation(s)
- Sahana Vasudevan
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Helma David
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Lakshmi Chanemougam
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Jayalakshmi Ramani
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Maanasa Ramesh Sangeetha
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| |
Collapse
|
20
|
Jiang JH, Cameron DR, Nethercott C, Aires-de-Sousa M, Peleg AY. Virulence attributes of successful methicillin-resistant Staphylococcus aureus lineages. Clin Microbiol Rev 2023; 36:e0014822. [PMID: 37982596 PMCID: PMC10732075 DOI: 10.1128/cmr.00148-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a leading cause of severe and often fatal infections. MRSA epidemics have occurred in waves, whereby a previously successful lineage has been replaced by a more fit and better adapted lineage. Selection pressures in both hospital and community settings are not uniform across the globe, which has resulted in geographically distinct epidemiology. This review focuses on the mechanisms that trigger the establishment and maintenance of current, dominant MRSA lineages across the globe. While the important role of antibiotic resistance will be mentioned throughout, factors which influence the capacity of S. aureus to colonize and cause disease within a host will be the primary focus of this review. We show that while MRSA possesses a diverse arsenal of toxins including alpha-toxin, the success of a lineage involves more than just producing toxins that damage the host. Success is often attributed to the acquisition or loss of genetic elements involved in colonization and niche adaptation such as the arginine catabolic mobile element, as well as the activity of regulatory systems, and shift metabolism accordingly (e.g., the accessory genome regulator, agr). Understanding exactly how specific MRSA clones cause prolonged epidemics may reveal targets for therapies, whereby both core (e.g., the alpha toxin) and acquired virulence factors (e.g., the Panton-Valentine leukocidin) may be nullified using anti-virulence strategies.
Collapse
Affiliation(s)
- Jhih-Hang Jiang
- Department of Microbiology, Infection Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - David R Cameron
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Cara Nethercott
- Department of Microbiology, Infection Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Marta Aires-de-Sousa
- Laboratory of Molecular Genetics, Institutode Tecnologia Químicae Biológica António Xavier (ITQB-NOVA), Universidade Nova de Lisboa, Oeiras, Portugal
- Escola Superior de Saúde da Cruz Vermelha Portuguesa-Lisboa (ESSCVP-Lisboa), Lisbon, Portugal
| | - Anton Y Peleg
- Department of Microbiology, Infection Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Centre to Impact Antimicrobial Resistance, Monash University, Clayton, Melbourne, Victoria, Australia
| |
Collapse
|
21
|
Singh TP, Farias Amorim C, Lovins VM, Bradley CW, Carvalho LP, Carvalho EM, Grice EA, Scott P. Regulatory T cells control Staphylococcus aureus and disease severity of cutaneous leishmaniasis. J Exp Med 2023; 220:e20230558. [PMID: 37812390 PMCID: PMC10561556 DOI: 10.1084/jem.20230558] [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: 04/03/2023] [Revised: 08/02/2023] [Accepted: 09/19/2023] [Indexed: 10/10/2023] Open
Abstract
Cutaneous leishmaniasis causes alterations in the skin microbiota, leading to pathologic immune responses and delayed healing. However, it is not known how these microbiota-driven immune responses are regulated. Here, we report that depletion of Foxp3+ regulatory T cells (Tregs) in Staphylococcus aureus-colonized mice resulted in less IL-17 and an IFN-γ-dependent skin inflammation with impaired S. aureus immunity. Similarly, reducing Tregs in S. aureus-colonized and Leishmania braziliensis-infected mice increased IFN-γ, S. aureus, and disease severity. Importantly, analysis of lesions from L. braziliensis patients revealed that low FOXP3 gene expression is associated with high IFNG expression, S. aureus burden, and delayed lesion resolution compared to patients with high FOXP3 expression. Thus, we found a critical role for Tregs in regulating the balance between IL-17 and IFN-γ in the skin, which influences both bacterial burden and disease. These results have clinical ramifications for cutaneous leishmaniasis and other skin diseases associated with a dysregulated microbiome when Tregs are limited or dysfunctional.
Collapse
Affiliation(s)
- Tej Pratap Singh
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Camila Farias Amorim
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Victoria M. Lovins
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Charles W. Bradley
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lucas P. Carvalho
- Servico de Imunologia, Complexo Hospitalar Universitario Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
- Laboratorio de Pesquisas Clinicas do Instituto de Pesquisas Goncalo Moniz, Fiocruz, Salvador, Brazil
| | - Edgar M. Carvalho
- Servico de Imunologia, Complexo Hospitalar Universitario Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
- Laboratorio de Pesquisas Clinicas do Instituto de Pesquisas Goncalo Moniz, Fiocruz, Salvador, Brazil
| | - Elizabeth A. Grice
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
22
|
Si W, Li M, Wang K, Li J, Xu M, Zhou X, Bai J, Qu Z, Song G, Wu X, Guo Y, Hu H, Fu D, Yang Z, Wu M, Yan D, Song X, Tian Z. Staphylococcus warneri strain XSB102 exacerbates psoriasis and promotes keratinocyte proliferation in imiquimod-induced psoriasis-like dermatitis mice. Arch Microbiol 2023; 206:3. [PMID: 37991548 DOI: 10.1007/s00203-023-03726-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/19/2023] [Accepted: 10/29/2023] [Indexed: 11/23/2023]
Abstract
Psoriasis is one of the common chronic inflammatory skin diseases worldwide. The skin microbiota plays a role in psoriasis through regulating skin homeostasis. However, the studies on the interactions between symbiotic microbial strains and psoriasis are limited. In this study, Staphylococcus strain XSB102 was isolated from the skin of human, which was identified as Staphylococcus warneri using VITEK2 Compact. To reveal the roles of Staphylococcus warneri on psoriasis, XSB102 were applied on the back of imiquimod-induced psoriasis-like dermatitis mice. The results indicated that it exacerbated the psoriasis and significantly increased the thickening of the epidermis. Furthermore, in vitro experiments confirmed that inactivated strain XSB102 could promote the proliferation of human epidermal keratinocytes (HaCaT) cell. However, real-time quantitative PCR and immunofluorescence results suggested that the expression of inflammatory factors such as IL-17a, IL-6, and so on were not significantly increased, while extracellular matrix related factors such as Col6a3 and TGIF2 were significantly increased after XSB102 administration. This study indicates that Staphylococcus warneri XSB102 can exacerbate psoriasis and promote keratinocyte proliferation independently of inflammatory factors, which paves the way for further exploration of the relationship between skin microbiota and psoriasis.
Collapse
Affiliation(s)
- Wenhao Si
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
- Department of Dermatology, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Min Li
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Kuan Wang
- Department of Dermatology, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Jialin Li
- Department of Dermatology, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Mengke Xu
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Xiaoyue Zhou
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Jie Bai
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Zhiyuan Qu
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Guoyan Song
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Xueya Wu
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Yuqi Guo
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Hua Hu
- Department of Dermatology, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Dandan Fu
- Department of Dermatology, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Zishan Yang
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Minna Wu
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Dong Yan
- Xinxiang Key Laboratory of Pathogenic Biology, Department of Pathogenic Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
| | - Xiangfeng Song
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
| | - Zhongwei Tian
- Department of Dermatology, the First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, Henan, China.
| |
Collapse
|
23
|
Zhang Y, Yang F, Sun D, Xu L, Shi Y, Qin L, Zhao L, Wang L, Sun W, Wu H, Lu D, Zhang W, Luo P, Cheng P, Zou Q, Zeng H. rFSAV promotes Staphylococcus aureus-infected bone defect healing via IL-13- mediated M2 macrophage polarization. Clin Immunol 2023; 255:109747. [PMID: 37634854 DOI: 10.1016/j.clim.2023.109747] [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: 06/23/2023] [Revised: 08/16/2023] [Accepted: 08/22/2023] [Indexed: 08/29/2023]
Abstract
Staphylococcus aureus (S. aureus) contamination commonly occurs in orthopedic internal fixation operations, leading to a delayed healing of the defected bone tissue. However, antibiotic treatments are ineffective in dealing with S. aureus bone infections due to the rise in multiple antimicrobial resistances. Here, we reported the protective effects of a recombinant five-antigen S. aureus vaccine (rFSAV) in an S. aureus infected bone defect model. In this study, we found the number of M2 macrophages markedly increased in the defect site and played a critical role in the healing of defected bone mediated by rFSAV. Mechanistically, rFSAV mediated increased level of IL-13 in bone defect site predominant M2 macrophage polarization. In summary, our study reveals a key role of M2 macrophage polarization in the bone regeneration process in S. aureus infection induced bone defect, which provide a promising application of rFSAV for the treatment of bone infection for orthopedic applications.
Collapse
Affiliation(s)
- Yanhao Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Feng Yang
- Chengdu Olymvax Biopharmaceuticals Inc., Chengdu, Sichuan 611731, PR China
| | - Dong Sun
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Limin Xu
- Chengdu Olymvax Biopharmaceuticals Inc., Chengdu, Sichuan 611731, PR China
| | - Yaojia Shi
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Leilei Qin
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400038, China
| | - Liqun Zhao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Liting Wang
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China
| | - Wei Sun
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China
| | - Hongri Wu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Dongshui Lu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Weijun Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Ping Luo
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Ping Cheng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China.
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, China; State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University, Chongqing, China.
| |
Collapse
|
24
|
Leyva-Castillo JM, McGurk A, Strakosha M, Vega-Mendoza D, Smith SEM, Stafstrom K, Elkins M, Chou J, Wang YH, Geha RS. IL-4 receptor alpha blockade dampens allergic inflammation and upregulates IL-17A expression to promote Saureus clearance in antigen sensitized mouse skin. J Allergy Clin Immunol 2023; 152:907-915. [PMID: 37315811 PMCID: PMC10592541 DOI: 10.1016/j.jaci.2023.05.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Skin colonization with Staphylococcus aureus aggravates atopic dermatitis and exaggerates allergic skin inflammation in mice. IL-4 receptor α (IL-4Rα) blockade is beneficial in atopic dermatitis and reduces Saureus skin colonization through unknown mechanisms. The cytokine IL-17A restrains Saureus growth. OBJECTIVES This study sought to examine the effect of IL-4Rα blockade on Saureus colonization at sites of allergic skin inflammation in mice and determine the mechanism involved. METHODS BALB/c mice were epicutaneously sensitized with ovalbumin (OVA). Immediately after, PSVue 794-labeled S aureus strain SF8300 or saline was applied and a single dose of anti-IL-4Rα blocking antibody, a mixture of anti-IL-4Rα and anti-IL-17A blocking antibodies, or IgG isotype controls were administered intradermally. Saureus load was assessed 2 days later by in vivo imaging and enumeration of colony forming units. Skin cellular infiltration was examined by flow cytometry and gene expression by quantitative PCR and transcriptome analysis. RESULTS IL-4Rα blockade decreased allergic skin inflammation in OVA-sensitized skin, as well as in OVA-sensitized and Saureus-exposed skin, evidenced by significantly decreased epidermal thickening and reduced dermal infiltration by eosinophils and mast cells. This was accompanied by increased cutaneous expression of Il17a and IL-17A-driven antimicrobial genes with no change in Il4 and Il13 expression. IL-4Rα blockade significantly decreased Saureus load in OVA-sensitized and S aureus-exposed skin. IL-17A blockade reversed the beneficial effect of IL-4Rα blockade on Saureus clearance and reduced the cutaneous expression of IL-17A-driven antimicrobial genes. CONCLUSIONS IL-4Rα blockade promotes Saureus clearance from sites of allergic skin inflammation in part by enhancing IL-17A expression.
Collapse
Affiliation(s)
| | - Alex McGurk
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Maria Strakosha
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Daniela Vega-Mendoza
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Sophia E M Smith
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Kelsey Stafstrom
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Megan Elkins
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Janet Chou
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | | | - Raif S Geha
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass.
| |
Collapse
|
25
|
Wang Y, Tang M, Deng H, Hong Z, Liang Z, Huang Y, Zeng C, Yang K. Ampelopsin attenuates Staphylococcus aureus Alpha-Toxin-Induced Lung Injury. Microb Pathog 2023; 183:106316. [PMID: 37634577 DOI: 10.1016/j.micpath.2023.106316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/18/2023] [Accepted: 08/20/2023] [Indexed: 08/29/2023]
Abstract
Staphylococcus aureus is a prevalent cause of lung infections in hospitals and communities, and can cause a wide spectrum of human infections. Due to the bottleneck caused by antibiotic resistance and substantial increases in morbidity and mortality, targeting the virulence factors released by S. aureus as an alternative prevention and treatment method has become a promising approach. Ampelopsin, a component of vine tea, has promising potential for treating S. aureus-induced acute lung injury. In this study, the effects of ampelopsin were investigated on a mouse model of acute lung injury established using S. aureus 8325-4 and the α-hemolysin (hla) silent strain DU1090. The hla silent strain did not cause mortality in mice, whereas lethal and sublethal concentrations of S. aureus 8325-4 caused high mortality. Notably, ampelopsin treatment protected against mortality stemming from S. aureus infection. Ampelopsin yielded enhancements in lung barrier function, decreased total protein leakage in the alveolar lavage fluid, and modulated inflammatory signaling pathway-related proteins, thereby reducing the release of pro-inflammatory factors and improving respiratory dysfunction. Moreover, ampelopsin prevented the upregulation of ADAM10 activity, leading to E-cadherin mucin cleavage. In conclusion, our findings establish the key role of alpha -toxin in infectious lung injury in S. aureus and provide support for ampelopsin as an effective therapeutic approach to improve lung injury.
Collapse
Affiliation(s)
- Yi Wang
- School of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530200, PR China
| | - Mulan Tang
- School of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530200, PR China
| | - Haojian Deng
- School of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530200, PR China
| | - Zhengshan Hong
- School of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530200, PR China
| | - Zhi Liang
- School of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530200, PR China
| | - Yumei Huang
- School of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530200, PR China
| | - Chunhui Zeng
- School of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530200, PR China.
| | - Ke Yang
- School of Pharmacy, Guangxi University of Chinese Medicine, Nanning, 530200, PR China.
| |
Collapse
|
26
|
Cela EM, Urquiza D, Gómez MI, Gonzalez CD. New Weapons to Fight against Staphylococcus aureus Skin Infections. Antibiotics (Basel) 2023; 12:1477. [PMID: 37887178 PMCID: PMC10603739 DOI: 10.3390/antibiotics12101477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/28/2023] Open
Abstract
The treatment of Staphylococcus aureus skin and soft tissue infections faces several challenges, such as the increased incidence of antibiotic-resistant strains and the fact that the antibiotics available to treat methicillin-resistant S. aureus present low bioavailability, are not easily metabolized, and cause severe secondary effects. Moreover, besides the susceptibility pattern of the S. aureus isolates detected in vitro, during patient treatment, the antibiotics may never encounter the bacteria because S. aureus hides within biofilms or inside eukaryotic cells. In addition, vascular compromise as well as other comorbidities of the patient may impede proper arrival to the skin when the antibiotic is given parenterally. In this manuscript, we revise some of the more promising strategies to improve antibiotic sensitivity, bioavailability, and delivery, including the combination of antibiotics with bactericidal nanomaterials, chemical inhibitors, antisense oligonucleotides, and lytic enzymes, among others. In addition, alternative non-antibiotic-based experimental therapies, including the delivery of antimicrobial peptides, bioactive glass nanoparticles or nanocrystalline cellulose, phototherapies, and hyperthermia, are also reviewed.
Collapse
Affiliation(s)
- Eliana M. Cela
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1425FQB, Argentina; (E.M.C.); (D.U.); (M.I.G.)
- Cátedra de Inmunología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires C1113AAD, Argentina
| | - Dolores Urquiza
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1425FQB, Argentina; (E.M.C.); (D.U.); (M.I.G.)
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Departamento de Investigaciones Biomédicas y Biotecnológicas, Universidad Maimónides, Buenos Aires C1405BCK, Argentina
| | - Marisa I. Gómez
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1425FQB, Argentina; (E.M.C.); (D.U.); (M.I.G.)
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Departamento de Investigaciones Biomédicas y Biotecnológicas, Universidad Maimónides, Buenos Aires C1405BCK, Argentina
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Cintia D. Gonzalez
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1425FQB, Argentina; (E.M.C.); (D.U.); (M.I.G.)
- Cátedra de Inmunología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires C1113AAD, Argentina
| |
Collapse
|
27
|
Dai X, Liu X, Li Y, Xu Q, Yang L, Gao F. Nitrogen-phosphorous co-doped carbonized chitosan nanoparticles for chemotherapy and ROS-mediated immunotherapy of intracellular Staphylococcus aureus infection. Carbohydr Polym 2023; 315:121013. [PMID: 37230629 DOI: 10.1016/j.carbpol.2023.121013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 05/07/2023] [Accepted: 05/08/2023] [Indexed: 05/27/2023]
Abstract
Staphylococcus aureus (S. aureus) residing in host macrophages is hard to clear because intracellular S. aureus has evolved mechanisms to hijack and subvert the immune response to favor intracellular infection. To overcome this challenge, nitrogen-phosphorous co-doped carbonized chitosan nanoparticles (NPCNs), which possess the polymer/carbon hybrid structures, were fabricated to clear intracellular S. aureus infection through chemotherapy and immunotherapy. Multi-heteroatom NPCNs were fabricated through the hydrothermal method, where chitosan and imidazole were used as the C and N sources and phosphoric acid as the P source. NPCNs can not only be used as a fluorescent probe for bacteria imaging but also kill extracellular and intracellular bacteria with low cytotoxicity. NPCNs could generate ROS and polarize macrophages into classically activated (M1) phenotypes to increase antibacterial immunity. Furthermore, NPCNs could accelerate intracellular S. aureus-infected wound healing in vivo. We envision that these carbonized chitosan nanoparticles may provide a new platform for clearing intracellular bacterial infection through chemotherapy and ROS-mediated immunotherapy.
Collapse
Affiliation(s)
- Xiaomei Dai
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Key Laboratory of Chemo/Biosensing, Laboratory of Biosensing and Bioimaging (LOBAB), College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, PR China.
| | - Xiaojun Liu
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Key Laboratory of Chemo/Biosensing, Laboratory of Biosensing and Bioimaging (LOBAB), College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, PR China
| | - Yu Li
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Key Laboratory of Chemo/Biosensing, Laboratory of Biosensing and Bioimaging (LOBAB), College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, PR China
| | - Qingqing Xu
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Key Laboratory of Chemo/Biosensing, Laboratory of Biosensing and Bioimaging (LOBAB), College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, PR China
| | - Lele Yang
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Key Laboratory of Chemo/Biosensing, Laboratory of Biosensing and Bioimaging (LOBAB), College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, PR China
| | - Feng Gao
- Laboratory of Functionalized Molecular Solids, Ministry of Education, Anhui Key Laboratory of Chemo/Biosensing, Laboratory of Biosensing and Bioimaging (LOBAB), College of Chemistry and Materials Science, Anhui Normal University, Wuhu 241002, PR China.
| |
Collapse
|
28
|
Battaglia M, Garrett-Sinha LA. Staphylococcus xylosus and Staphylococcus aureus as commensals and pathogens on murine skin. Lab Anim Res 2023; 39:18. [PMID: 37533118 PMCID: PMC10394794 DOI: 10.1186/s42826-023-00169-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023] Open
Abstract
Skin ulcers, skin dermatitis and skin infections are common phenomena in colonies of laboratory mice and are often found at increased prevalence in certain immunocompromised strains. While in many cases these skin conditions are mild, in other cases they can be severe and lead to animal morbidity. Furthermore, the presence of skin infections and ulcerations can complicate the interpretation of experimental protocols, including those examining immune cell activation. Bacterial species in the genus Staphylococcus are the most common pathogens recovered from skin lesions in mice. In particular, Staphylococcus aureus and Staphylococcus xylosus have both been implicated as pathogens on murine skin. Staphylococcus aureus is a well-known pathogen of human skin, but S. xylosus skin infections in humans have not been described, indicating that there is a species-specific difference in the ability of S. xylosus to serve as a skin pathogen. The aim of this review is to summarize studies that link S. aureus and S. xylosus to skin infections of mice and to describe factors involved in their adherence to tissue and their virulence. We discuss potential differences in mouse and human skin that might underlie the ability of S. xylosus to act as a pathogen on murine skin, but not human skin. Finally, we also describe mouse mutants that have shown increased susceptibility to skin infections with staphylococcal bacteria. These mutants point to pathways that are important in the control of commensal staphylococcal bacteria. The information here may be useful to researchers who are working with mouse strains that are prone to skin infections with staphylococcal bacteria.
Collapse
Affiliation(s)
- Michael Battaglia
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Lee Ann Garrett-Sinha
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA.
| |
Collapse
|
29
|
Elmaidomy AH, Mohamad SA, Abdelnaser M, Yahia R, Mokhtar FA, Alsenani F, Badr MY, Almaghrabi SY, Altemani FH, Alzubaidi MA, Saber EA, Elrehany MA, Abdelmohsen UR, Sayed AM. Vitis vinifera leaf extract liposomal Carbopol gel preparation's potential wound healing and antibacterial benefits: in vivo, phytochemical, and computational investigation. Food Funct 2023; 14:7156-7175. [PMID: 37462414 DOI: 10.1039/d2fo03212k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Vitis vinifera Egyptian edible leaf extract loaded on a soybean lecithin, cholesterol, and Carbopol gel preparation (VVL-liposomal gel) was prepared to maximize the in vivo wound healing and anti-MRSA activities for the crude extract, using an excision wound model and focusing on TLR-2, MCP-1, CXCL-1, CXCL-2, IL-6 and IL-1β, and MRSA (wound infection model, and peritonitis infection model). VVL-liposomal gel was stable with significant drug entrapment efficiency reaching 88% ± 3, zeta potential value ranging from -50 to -63, and a size range of 50-200 μm nm in diameter. The in vivo evaluation proved the ability of VVL-liposomal gel to gradually release the drugs in a sustained manner with greater complete wound healing effect and tissue repair after 7 days of administration, with a significant decrease in bacterial count compared with the crude extract. Phytochemical investigation of the crude extract of the leaves yielded fourteen compounds: two new stilbenes (1, 2), along with twelve known ones (3-14). Furthermore, a computational study was conducted to identify the genes and possible pathways responsible for the anti-MRSA activity of the isolated compounds, and inverse docking was used to identify the most likely molecular targets that could mediate the extract's antibacterial activity. Gyr-B was discovered to be the best target for compounds 1 and 2. Hence, VVL-liposomal gel can be used as a novel anti-dermatophytic agent with potent wound healing and anti-MRSA capacity, paving the way for future clinical research.
Collapse
Affiliation(s)
- Abeer H Elmaidomy
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt.
| | - Soad A Mohamad
- Department of Pharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, Deraya University, New Minya 61111, Egypt
| | - Mahmoud Abdelnaser
- Department of Biochemistry, Faculty of Pharmacy, Deraya University, New Minya 61111, Egypt
| | - Ramadan Yahia
- Department of Microbiology and Immunology, Faculty of Pharmacy, Deraya University, Universities Zone, New Minya City 61111, Egypt
| | - Fatma A Mokhtar
- Department of Pharmacognosy, Faculty of Pharmacy, El Saleheya El Gadida University, El Saleheya El Gadida 44813, Sharkia, Egypt
| | - Faisal Alsenani
- Department of Pharmacognosy, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia.
| | - Moutaz Y Badr
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah 24381, Saudi Arabia.
| | - Safa Y Almaghrabi
- Department of Physiology, Faculty of Medicine, King Abduaziz University, Jeddah 22252, Saudi Arabia.
| | - Faisal H Altemani
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Mubarak A Alzubaidi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Entesar Ali Saber
- Department of Histology and Cell Biology, Faculty of Medicine, Minia University, Minya 61519, Egypt
- Delegated to Deraya University, Universities Zone, New Minya 61111, Egypt
| | - Mahmoud A Elrehany
- Department of Biochemistry, Faculty of Pharmacy, Deraya University, New Minya 61111, Egypt
| | - Usama Ramadan Abdelmohsen
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minya 61519, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Deraya University, New Minya 61111, Egypt
| | - Ahmed M Sayed
- Department of Pharmacognosy, Faculty of Pharmacy, Nahda University, Beni-Suef 62513, Egypt.
| |
Collapse
|
30
|
Toller-Kawahisa JE, Hiroki CH, Silva CMDS, Nascimento DC, Públio GA, Martins TV, Damasceno LEA, Veras FP, Viacava PR, Sukesada FY, Day EA, Zotta A, Ryan TAJ, Moreira da Silva R, Cunha TM, Lopes NP, Cunha FDQ, O'Neill LAJ, Alves-Filho JC. The metabolic function of pyruvate kinase M2 regulates reactive oxygen species production and microbial killing by neutrophils. Nat Commun 2023; 14:4280. [PMID: 37460614 DOI: 10.1038/s41467-023-40021-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/03/2023] [Indexed: 07/20/2023] Open
Abstract
Neutrophils rely predominantly on glycolytic metabolism for their biological functions, including reactive oxygen species (ROS) production. Although pyruvate kinase M2 (PKM2) is a glycolytic enzyme known to be involved in metabolic reprogramming and gene transcription in many immune cell types, its role in neutrophils remains poorly understood. Here, we report that PKM2 regulates ROS production and microbial killing by neutrophils. Zymosan-activated neutrophils showed increased cytoplasmic expression of PKM2. Pharmacological inhibition or genetic deficiency of PKM2 in neutrophils reduced ROS production and Staphylococcus aureus killing in vitro. In addition, this also resulted in phosphoenolpyruvate (PEP) accumulation and decreased dihydroxyacetone phosphate (DHAP) production, which is required for de novo synthesis of diacylglycerol (DAG) from glycolysis. In vivo, PKM2 deficiency in myeloid cells impaired the control of infection with Staphylococcus aureus. Our results fill the gap in the current knowledge of the importance of lower glycolysis for ROS production in neutrophils, highlighting the role of PKM2 in regulating the DHAP and DAG synthesis to promote ROS production in neutrophils.
Collapse
Affiliation(s)
- Juliana Escher Toller-Kawahisa
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
| | - Carlos Hiroji Hiroki
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Camila Meirelles de Souza Silva
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Daniele Carvalho Nascimento
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Gabriel Azevedo Públio
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Timna Varela Martins
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Luis Eduardo Alves Damasceno
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Flávio Protásio Veras
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Paula Ramos Viacava
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Fábio Yuji Sukesada
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Emily Anne Day
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Alessia Zotta
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | | | - Rodrigo Moreira da Silva
- NPPNS, Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirao Preto, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Norberto Peporine Lopes
- NPPNS, Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirao Preto, Brazil
| | - Fernando de Queiroz Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Luke Anthony John O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - José Carlos Alves-Filho
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
- Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
| |
Collapse
|
31
|
Rancan F, Jurisch J, Hadam S, Vogt A, Blume-Peytavi U, Bayer IS, Contardi M, Schaudinn C. Ciprofloxacin-Loaded Polyvinylpyrrolidone Foils for the Topical Treatment of Wound Infections with Methicillin-Resistant Staphylococcus aureus (MRSA). Pharmaceutics 2023; 15:1876. [PMID: 37514062 PMCID: PMC10385417 DOI: 10.3390/pharmaceutics15071876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/23/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Bacterial infections are a constant challenge in the management of acute and chronic wounds. Chronic wounds, such as diabetic foot ulcers, have increased significantly in the last few years due to the rise of an aging population. A better understanding of the infectious pathophysiological mechanisms is urgently needed along with new options for the treatment of wound infections and wound-healing disorders. New advances in the preparation of biocompatible dressing materials that can be loaded with antimicrobial drugs may improve the topical treatment of infected wounds. In this study, we investigated the antimicrobial activity of polyvinylpyrrolidone (PVP) foils loaded with ciprofloxacin (Cipro-foils) in the presence of acetic acid as a co-solvent. We used ex vivo human wounds that were infected with two bacterial strains: methicillin-resistant Staphylococcus aureus (MRSA) or Pseudomonas aeruginosa (PAO1). The effectiveness of the treatment was demonstrated by the quantification of the living bacteria extracted from the wound and the detection of released immunological mediators in skin extracts and in the skin culture media. We found that Cipro-foils effectively treated the infection with both PAO1 and MRSA. Other than PAO1, MRSA had no lytic activity toward skin proteins. MRSA infections increased cytokines' expression and release. Interestingly, treatment with Cipro-foils could partially counteract these effects.
Collapse
Affiliation(s)
- Fiorenza Rancan
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Jana Jurisch
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Sabrina Hadam
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Annika Vogt
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Ulrike Blume-Peytavi
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Ilker S Bayer
- Smart Materials, Istituto Italiano di Tecnologia, 16163 Genova, Italy
| | - Marco Contardi
- Smart Materials, Istituto Italiano di Tecnologia, 16163 Genova, Italy
- Department of Earth and Environmental Sciences (DISAT), University of Milan-Bicocca, Piazza della Scienza, 20126 Milan, Italy
| | - Christoph Schaudinn
- Advanced Light and Electron Microscopy, Zentrum für Biologische Gefahren und Spezielle Pathogene 4, Robert Koch Institute, 13353 Berlin, Germany
| |
Collapse
|
32
|
Shen P, Zheng L, Qin X, Li D, Zhang Z, Zhao J, Lin H, Hong H, Zhou Z, Wu Z. Synthesis of structure-defined β-1,4-GlcNAc-modified wall teichoic acids as potential vaccine against methicillin-resistant Staphylococcus aureus. Eur J Med Chem 2023; 258:115553. [PMID: 37336068 DOI: 10.1016/j.ejmech.2023.115553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 06/08/2023] [Accepted: 06/08/2023] [Indexed: 06/21/2023]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a high priority pathogen due to its life-threating infections to human health. Development of prophylactic or therapeutic anti-MRSA vaccine is a potential approach to treat S. aureus infections and overcome the resistance crisis. β-1,4-GlcNAc glycosylated wall teichoic acids (WTAs) derived from S. aureus are a new type of antigen that is closely associated with β-lactam resistance. In this study, structure-defined β-1,4-GlcNAc-modified WTAs varied in chain length and numbers of GlcNAc modification were synthesized by an ionic liquid-supported oligosaccharide synthesis (ILSOS) strategy in high efficiency and chromatography-free approach. Then the obtained WTAs were conjugated with tetanus toxin (TT) as vaccine candidates and were further evaluated in a mouse model to determine the structure-immunogenicity relationship. In vivo immunological studies revealed that the WTAs-TT conjugates provoked robust T cell-dependent responses and elicited high levels of specific anti-WTAs IgG antibodies production associated with the WTAs structure including chain length as well as the β-1,4-GlcNAc modification pattern. Heptamer WTAs conjugate T6, carrying three copy of β-1,4-GlcNAc modified RboP, was identified to elicit the highest titers of specific antibody production. The T6 antisera exhibited the highest recognition and binding affinity and the most potent OP-killing activities to MSSA and MRSA cells. This study demonstrated that β-1,4-GlcNAc glycosylated WTAs are promising antigens for further development against MRSA.
Collapse
Affiliation(s)
- Peng Shen
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Lele Zheng
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Xinfang Qin
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Dan Li
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Zijiang Zhang
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Jie Zhao
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Han Lin
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Haofei Hong
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Zhifang Zhou
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Zhimeng Wu
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
33
|
Chatuphonprasert W, Tatiya-aphiradee N, Sutthanut K, Thammawat S, Puthongking P, Nopwinyoowong N, Jarukamjorn K. Combinatory effects of Dipterocarpus alatus twig emulgel: Wound-restoring, antibacterial, and anti-inflammatory activities against methicillin-resistant Staphylococcus aureus-infected mouse superficial wounds. Heliyon 2023; 9:e17483. [PMID: 37416687 PMCID: PMC10320117 DOI: 10.1016/j.heliyon.2023.e17483] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/22/2023] [Accepted: 06/20/2023] [Indexed: 07/08/2023] Open
Abstract
Dipterocarpus alatus has been used for the treatment of infectious skin diseases and ulcerative wounds in Thai traditional medicine. A major pathogen in human superficial skin infections is methicillin-resistant Staphylococcus aureus (MRSA). This study determined the wound healing, antibacterial, and anti-inflammatory activities of D. alatus twig emulgel against MRSA-infected mouse superficial skin wounds. Ethyl acetate-methanol crude extract of D. alatus twig was incorporated into emulgel at concentrations of 20 and 40 mg/g (D20 and D40) and its activity was compared to tetracycline emulgel (160 μg/g, Tetra). MRSA-infected superficial wounds demonstrated decreased skin barrier strength, increased transepidermal water loss (TEWL), and mast cell accumulation. Expression of toll-like receptor 2 (TLR-2), NF-κβ, TNFα, IL-1β, IL-6 and IL-10 genes were induced after MRSA infection. Daily application of 100 μL of D20 or D40 for 9 days restored skin barrier strength and TEWL while reducing mast cell and MRSA numbers compared to the non-treated group (MRSA-NT). The wounds treated with D20 and D40 were entirely healed on day 9. Expression of TLR-2 and cytokine-related genes NF-κβ, TNFα, IL-1β, IL-6 and IL-10 were normalized by treatment with either D20 or D40. Therefore, emulgel containing 20 to 40 mg/g ethyl acetate-methanol crude D. alatus twig extract is a good candidate for development as a topical formulation for MRSA-infected ulcerated wounds.
Collapse
Affiliation(s)
- Waranya Chatuphonprasert
- Division of Pre-clinic, Faculty of Medicine, Mahasarakham University, Maha Sarakham, 44000, Thailand
- Research Group for Pharmaceutical Activities of Natural Products Using Pharmaceutical Biotechnology (PANPB), Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Nitima Tatiya-aphiradee
- Research Group for Pharmaceutical Activities of Natural Products Using Pharmaceutical Biotechnology (PANPB), Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Khaetthareeya Sutthanut
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002 Thailand
| | - Sutthiwan Thammawat
- Division of Pre-clinic, Faculty of Medicine, Mahasarakham University, Maha Sarakham, 44000, Thailand
| | - Ploenthip Puthongking
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002 Thailand
| | - Naroeporn Nopwinyoowong
- Research Group for Pharmaceutical Activities of Natural Products Using Pharmaceutical Biotechnology (PANPB), Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Kanokwan Jarukamjorn
- Research Group for Pharmaceutical Activities of Natural Products Using Pharmaceutical Biotechnology (PANPB), Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002 Thailand
| |
Collapse
|
34
|
Darbouret- Hervier A, Assi N, Asensio MJ, Bernabe B, Lechevallier A, Iantomasi R, Rokbi B, Botelho-Nevers E, Ruiz S. Anti-staphylococcus aureus adaptive immunity is impaired in end-stage renal disease patients on hemodialysis: one-year longitudinal study. Front Immunol 2023; 14:1123160. [PMID: 37304264 PMCID: PMC10250961 DOI: 10.3389/fimmu.2023.1123160] [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: 12/13/2022] [Accepted: 03/27/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction Patients with end-stage renal disease (ESRD) display defects in adaptive and innate immunity, increasing susceptibility to infection. Staphylococcus aureus (S. aureus) is a major cause of bacteraemia in this population and is associated with increased mortality. More information on the immune response to S. aureus in these patients is needed to inform effective vaccine development. Methods A longitudinal prospective study was carried out at two medical centers and included 48 ESRD patients who started chronic hemodialysis (HD) treatment ≤3 months before inclusion. Control samples were taken from 62 consenting healthy blood donors. Blood samples were obtained from ESRD patients at each visit, on month (M) 0 (beginning of HD), M6 and M12. Around 50 immunological markers of adaptive and innate immunity were assessed to compare immune responses to S. aureus in ESRD patients versus controls to document the changes on their immune profile during HD. Results S. aureus survival in whole blood was significantly higher in ESRD patients than in controls at M0 (P=0.049), while impaired oxidative burst activity was observed in ESRD patients at all timepoints (P<0.001). S. aureus-specific immunoglobulin G (IgG) responses to iron surface determinant B (IsdB) and S. aureus α hemolysin (Hla) antigens were lower in ESRD patients than in healthy donors at M0 (P=0.003 and P=0.007, respectively) and M6 (P=0.05 and P=0.03, respectively), but were restored to control levels at M12. Moreover, S. aureus-specific T-helper cell responses were comparable to controls for IsdB but were impaired for Hla antigen at all timepoints: 10% of ESRD patients responded to Hla at M0, increasing to 30% at M12, compared with 45% of healthy donors. B-cell and T-cell concentrations in blood were significantly reduced (by 60% and 40%, respectively) compared with healthy controls. Finally, upregulation of Human Leucocyte Antigen-DR (HLA-DR) and C-C chemokine Receptor type 2 (CCR2) was impaired at M0 but was restored during the first year of HD. Conclusion All together, these results show that adaptive immunity was largely impaired in ESRD patients, whereas innate immunity was less impacted and tended to be restored by HD.
Collapse
Affiliation(s)
| | - Nada Assi
- Research Department, Sanofi, Marcy l’Etoile, France
| | | | | | | | | | - Bachra Rokbi
- Research Department, Sanofi, Marcy l’Etoile, France
| | - Elisabeth Botelho-Nevers
- Infectious Diseases Department, University Hospital, Saint-Etienne, France
- CIC Inserm, University Hospital, Saint-Etienne, France
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP, University, Lyon, Université Jean Monnet, Inserm, CNRS, Saint-Etienne, France
| | - Sophie Ruiz
- Research Department, Sanofi, Marcy l’Etoile, France
| |
Collapse
|
35
|
Zhao Y, Zhang Z, Ning Y, Miao P, Li Z, Wang H. Simultaneous quantitative analysis of Escherichia coli, Staphylococcus aureus and Salmonella typhimurium using surface-enhanced Raman spectroscopy coupled with partial least squares regression and artificial neural networks. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 293:122510. [PMID: 36812753 DOI: 10.1016/j.saa.2023.122510] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/12/2023] [Accepted: 02/14/2023] [Indexed: 06/18/2023]
Abstract
Simultaneous detection of mixed bacteria accurately and sensitively is a major challenge in microbial quality control field. In this study, we proposed a label-free SERS technique coupled with partial least squares regression (PLSR) and artificial neural networks (ANNs) for quantitative analysis of Escherichia coli, Staphylococcus aureus and Salmonella typhimurium simultaneously. SERS-active and reproducible Raman spectra can be acquired directly upon the bacteria and Au@Ag@SiO2 nanoparticle composites on the surface of gold foil substrates. After applying different preprocessing models, SERS-PLSR and SERS-ANNs quantitative analysis models were developed to map SERS spectra of concentrations of the Escherichia coli, Staphylococcus aureus and Salmonella typhimurium, respectively. Both models achieved high prediction accuracy and low prediction error, while the performance of SERS-ANNs model in both quality of fit (R2 > 0.95) and accuracy of predictions (RMSE < 0.06) was superior to SERS-PLSR model. Therefore, it is feasible to develop simultaneous quantitative analysis of mixed pathogenic bacteria by proposed SERS methodology.
Collapse
Affiliation(s)
- Yuwen Zhao
- College of Pharmaceutical Engineering of Traditional Chinese Medicine (TCM), Tianjin University of TCM, Tianjin 301617, China
| | - Zeshuai Zhang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine (TCM), Tianjin University of TCM, Tianjin 301617, China
| | - Ying Ning
- College of Pharmaceutical Engineering of Traditional Chinese Medicine (TCM), Tianjin University of TCM, Tianjin 301617, China
| | - Peiqi Miao
- Tianjin Modern Innovative TCM Technology Co., Ltd., Tianjin 300392, China
| | - Zheng Li
- College of Pharmaceutical Engineering of Traditional Chinese Medicine (TCM), Tianjin University of TCM, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of TCM, Tianjin 301617, China.
| | - Haixia Wang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine (TCM), Tianjin University of TCM, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of TCM, Tianjin 301617, China.
| |
Collapse
|
36
|
Yudasari N, Hardiansyah A, Herbani Y, Isnaeni, Suliyanti MM, Djuhana D. Single-step laser ablation synthesis of ZnO–Ag nanocomposites for broad-spectrum dye photodegradation and bacterial photoinactivation. J Photochem Photobiol A Chem 2023. [DOI: 10.1016/j.jphotochem.2023.114717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
37
|
Role of PI3K-AKT Pathway in Ultraviolet Ray and Hydrogen Peroxide-Induced Oxidative Damage and Its Repair by Grain Ferments. Foods 2023; 12:foods12040806. [PMID: 36832881 PMCID: PMC9957031 DOI: 10.3390/foods12040806] [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: 12/11/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/16/2023] Open
Abstract
UV and external environmental stimuli can cause oxidative damage to skin cells. However, the molecular mechanisms involved in cell damage have not been systematically and clearly elucidated. In our study, an RNA-seq technique was used to determine the differentially expressed genes (DEGs) of the UVA/H2O2-induced model. Gene Oncology (GO) clustering and the Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathway analysis were performed to determine the core DEGs and key signaling pathway. The PI3K-AKT signaling pathway was selected as playing a part in the oxidative process and was verified by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). We selected three kinds of Schizophyllum commune fermented actives to evaluate whether the PI3K-AKT signaling pathway also plays a role in the resistance of active substances to oxidative damage. Results indicated that DEGs were mainly enriched in five categories: external stimulus response, oxidative stress, immunity, inflammation, and skin barrier regulation. S. commune-grain ferments can effectively reduce cellular oxidative damage through the PI3K-AKT pathway at both the cellular and molecular levels. Some typical mRNAs (COL1A1, COL1A2, COL4A5, FN1, IGF2, NR4A1, and PIK3R1) were detected, and the results obtained were consistent with those of RNA-seq. These results may give us a common set of standards or criteria for the screen of anti-oxidative actives in the future.
Collapse
|
38
|
Scutera S, Sparti R, Comini S, Menotti F, Musso T, Cuffini AM, Allizond V, Banche G. Dalbavancin Boosts the Ability of Neutrophils to Fight Methicillin-Resistant Staphylococcus aureus. Int J Mol Sci 2023; 24:ijms24032541. [PMID: 36768864 PMCID: PMC9917267 DOI: 10.3390/ijms24032541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/18/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Polymorphonuclear leukocytes (PMNs) are the most important cell type involved in the early nonspecific host response to bacterial pathogens. Staphylococcus aureus has evolved mechanisms to evade immune responses that contribute to its persistence in PMNs, and acquired resistance to several antimicrobials. Additionally, methicillin-resistant S. aureus (MRSA) is one of the most common causes of acute bacterial skin and skin-structure infections (ABSSSIs). Dalbavancin (DBV), a lipoglycopeptide, is indicated for the treatment of ABSSSIs, and has a broad spectrum of action against most microorganisms. Here, we sought to determine the effect of DBV on the neutrophil killing of MRSA and its potential immunomodulating activity. Our results revealed that DBV boosts MRSA killing by acting on both bacteria and PMNs. DBV pre-treatment of PMNs did not change the respiratory burst or degranulation, while an increased trend in neutrophil extracellular traps-associated elastase and in the production of TNFα and CXCL8 was revealed. In parallel, DBV caused a delay in the apoptosis of MRSA-infected neutrophils. In conclusion, we demonstrated a cooperative effect between the antimicrobial properties of PMNs and DBV, thus owing to their immunomodulatory activity. In the choice of the treatment management of serious S. aureus infections, DBV should be considered as an outstanding option since it reinforces PMNs pathogen clearance capability by exerting its effect directly, not only on MRSA but also on neutrophils.
Collapse
|
39
|
Gram-Positive Bacteria Cell Wall Peptidoglycan Polymers Activate Human Dendritic Cells to Produce IL-23 and IL-1β and Promote T H17 Cell Differentiation. Microorganisms 2023; 11:microorganisms11010173. [PMID: 36677464 PMCID: PMC9867153 DOI: 10.3390/microorganisms11010173] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/22/2022] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
Gram-positive bacterial infections are a major cause of organ failure and mortality in sepsis. Cell wall peptidoglycan (PGN) is shed during bacterial replication, and Bacillus anthracis PGN promotes a sepsis-like pathology in baboons. Herein, we determined the ability of polymeric Bacillus anthracis PGN free from TLR ligands to shape human dendritic cell (DC) responses that are important for the initiation of T cell immunity. Monocyte-derived DCs from healthy donors were incubated with PGN polymers isolated from Bacillus anthracis and Staphylococcus aureus. PGN activated the human DCs, as judged by the increased expression of surface HLA-DR, CD83, the T cell costimulatory molecules CD40 and CD86, and the chemokine receptor CCR7. PGN elicited the DC production of IL-23, IL-6, and IL-1β but not IL-12p70. The PGN-stimulated DCs induced the differentiation of naïve allogeneic CD4+ T cells into T helper (TH) cells producing IL-17 and IL-21. Notably, the DCs from a subset of donors did not produce significant levels of IL-23 and IL-1β upon PGN stimulation, suggesting that common polymorphisms in immune response genes regulate the PGN response. In sum, purified PGN is a highly stimulatory cell wall component that activates human DCs to secrete proinflammatory cytokines and promote the differentiation of TH17 cells that are important for neutrophil recruitment in extracellular bacterial infections.
Collapse
|
40
|
Arooj M, Rehman A, Hyun CL, Rafique A, Kang HK, Hyun JW, Koh YS. Diesel exhaust particulate matter impairs Toll-like receptor signaling and host defense against staphylococcal cutaneous infection in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114443. [PMID: 38321662 DOI: 10.1016/j.ecoenv.2022.114443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 02/08/2024]
Abstract
Air pollution is an emerging cause of mortality, affecting nearly 5 million people each year. Exposure to diesel exhaust fine particulate matter (PM2.5) aggravates respiratory and skin conditions. However, its impact on the protective immunity of the skin remains poorly understood. This study aimed to investigate the underlying molecular mechanism for adverse effects of PM2.5 on the host protective immunity using in vitro cell and in vivo mouse model. Intracellular translocation of Toll-like receptor 9 (TLR9) and CpG-DNA internalization were assessed in dendritic cells without or with PM2.5 treatment using immunofluorescence staining. Cytokine and nitric oxide production were measured in dendritic cells and macrophages without or with PM2.5 treatment. NF-κB and MAPK signaling was determined using western blotting. Skin disease severity, bacterial loads, and cytokine production were assessed in cutaneous Staphylococcus aureus (S. aureus) infection mouse model. PM2.5 interfered with TLR9 activation by inhibiting both TLR9 trafficking to early endosomes and CpG-DNA internalization via clathrin-mediated endocytosis. In addition, exposure to PM2.5 inhibited various TLR-mediated nitric oxide and cytokine production as well as MAPK and NF-κB signaling. PM2.5 rendered mice more susceptible to staphylococcal skin infections. Our results suggest that exposure to PM impairs TLR signaling and dampens the host defense against staphylococcal skin infections. Our data provide a novel perspective into the impact of PM on protective immunity which is paramount to revealing air pollutant-mediated toxicity on the host immunity.
Collapse
Affiliation(s)
- Madeeha Arooj
- College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, 102 Jejudaehakno, Jeju 63243, South Korea
| | - Abdur Rehman
- College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, 102 Jejudaehakno, Jeju 63243, South Korea
| | - Chang Lim Hyun
- College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, 102 Jejudaehakno, Jeju 63243, South Korea
| | - Asma Rafique
- College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, 102 Jejudaehakno, Jeju 63243, South Korea
| | - Hee-Kyoung Kang
- College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, 102 Jejudaehakno, Jeju 63243, South Korea
| | - Jin Won Hyun
- College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, 102 Jejudaehakno, Jeju 63243, South Korea
| | - Young-Sang Koh
- College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, 102 Jejudaehakno, Jeju 63243, South Korea.
| |
Collapse
|
41
|
Microbial Natural Products with Wound-Healing Properties. Processes (Basel) 2022. [DOI: 10.3390/pr11010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Wound healing continues to pose a challenge in clinical settings. Moreover, wound management must be performed properly and efficiently. Acute wound healing involves multiple cell divisions, a new extracellular matrix, and the process of formation, such as growth factors and cytokines, which are released at the site of the wound to regulate the process. Any changes that disrupt the healing process could cause tissue damage and prolong the healing process. Various factors, such as microbial infection, oxidation, and inflammation, can delay wound healing. In order to counter these problems, utilizing natural products with wound-healing effects has been reported to promote this process. Several natural products have been associated with wound healing, most of which are from medicinal plants. However, secondary microbial metabolites have not been extensively studied for their wound-healing properties. Further, investigations on the wound-healing control of natural microbial products are required due to a lack of studies. This review discussed the in vivo and in vitro research on the wound healing activities of natural microbial products, which may assist in the development of better wound treatments in the future.
Collapse
|
42
|
Hydrogels with intrinsic antibacterial activity prepared from naphthyl anthranilamide (NaA) capped peptide mimics. Sci Rep 2022; 12:22259. [PMID: 36564414 PMCID: PMC9789043 DOI: 10.1038/s41598-022-26426-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
In this study, we prepared antibacterial hydrogels through the self-assembly of naphthyl anthranilamide (NaA) capped amino acid based cationic peptide mimics. These ultra-short cationic peptide mimics were rationally designed with NaA as a capping group, L-phenylalanine, a short aliphatic linker, and a cationic group. The synthesized peptide mimics efficiently formed hydrogels with minimum gel concentrations between 0.1 and 0.3%w/v. The resulting hydrogels exhibited desirable viscoelastic properties which can be tuned by varying the cationic group, electronegative substituent, or counter anion. Importantly, nanofibers from the NaA-capped cationic hydrogels were found to be the source of hydrogels' potent bacteriacidal actvity against both Gram-positive and Gram-negative bacteria while remaining non-cytotoxic. These intrinsically antibacterial hydrogels are ideal candidates for further development in applications where bacterial contamination is problematic.
Collapse
|
43
|
Cavagnero KJ, Gallo RL. Essential immune functions of fibroblasts in innate host defense. Front Immunol 2022; 13:1058862. [PMID: 36591258 PMCID: PMC9797514 DOI: 10.3389/fimmu.2022.1058862] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/09/2022] [Indexed: 12/23/2022] Open
Abstract
The term fibroblast has been used generally to describe spindle-shaped stromal cells of mesenchymal origin that produce extracellular matrix, establish tissue structure, and form scar. Current evidence has found that cells with this morphology are highly heterogeneous with some fibroblastic cells actively participating in both innate and adaptive immune defense. Detailed analysis of barrier tissues such as skin, gut, and lung now show that some fibroblasts directly sense pathogens and other danger signals to elicit host defense functions including antimicrobial activity, leukocyte recruitment, and production of cytokines and lipid mediators relevant to inflammation and immunosuppression. This review will synthesize current literature focused on the innate immune functions performed by fibroblasts at barrier tissues to highlight the previously unappreciated importance of these cells in immunity.
Collapse
Affiliation(s)
| | - Richard L. Gallo
- Department of Dermatology, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
44
|
Kratofil RM, Kubes P. Recruited monocytes repair infections. Clin Transl Med 2022; 12:e1121. [PMID: 36424766 PMCID: PMC9691900 DOI: 10.1002/ctm2.1121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/27/2022] Open
Affiliation(s)
- Rachel M. Kratofil
- Department of Physiology and PharmacologyUniversity of CalgaryCalgaryAlbertaCanada
| | - Paul Kubes
- Calvin, Phoebe, and Joan Snyder Institute for Chronic DiseasesUniversity of CalgaryCalgaryAlbertaCanada
| |
Collapse
|
45
|
Rizzetto G, Molinelli E, Radi G, Cirioni O, Brescini L, Giacometti A, Offidani A, Simonetti O. MRSA and Skin Infections in Psoriatic Patients: Therapeutic Options and New Perspectives. Antibiotics (Basel) 2022; 11:1504. [PMID: 36358159 PMCID: PMC9686594 DOI: 10.3390/antibiotics11111504] [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: 09/23/2022] [Revised: 10/23/2022] [Accepted: 10/27/2022] [Indexed: 09/29/2023] Open
Abstract
Psoriatic patients present various infectious risk factors, but there are few studies in the literature evaluating the actual impact of psoriasis in severe staphylococcal skin infections. Our narrative review of the literature suggests that psoriatic patients are at increased risk of both colonization and severe infection, during hospitalization, by S. aureus. The latter also appears to play a role in the pathogenesis of psoriasis through the production of exotoxins. Hospitalized psoriatic patients are also at increased risk of MRSA skin infections. For this reason, new molecules are needed that could both overcome bacterial resistance and inhibit exotoxin production. In our opinion, in the near future, topical quorum sensing inhibitors in combination with current anti-MRSA therapies will be able to overcome the increasing resistance and block exotoxin production. Supplementation with Vitamin E (VE) or derivatives could also enhance the effect of anti-MRSA antibiotics, considering that psoriatic patients with metabolic comorbidities show a low intake of VE and low serum levels, making VE supplementation an interesting new perspective.
Collapse
Affiliation(s)
- Giulio Rizzetto
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Elisa Molinelli
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Giulia Radi
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Oscar Cirioni
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Lucia Brescini
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Andrea Giacometti
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Annamaria Offidani
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Oriana Simonetti
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| |
Collapse
|
46
|
Lecron JC, Charreau S, Jégou JF, Salhi N, Petit-Paris I, Guignouard E, Burucoa C, Favot-Laforge L, Bodet C, Barra A, Huguier V, Mcheik J, Dumoutier L, Garnier J, Bernard FX, Ryffel B, Morel F. IL-17 and IL-22 are pivotal cytokines to delay wound healing of S. aureus and P. aeruginosa infected skin. Front Immunol 2022; 13:984016. [PMID: 36275755 PMCID: PMC9585169 DOI: 10.3389/fimmu.2022.984016] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
IntroductionAlthough the presence of pathogens in skin wounds is known to delay the wound healing process, the mechanisms underlying this delay remain poorly understood. In the present study, we have investigated the regulatory role of proinflammatory cytokines on the healing kinetics of infected wounds.MethodsWe have developed a mouse model of cutaneous wound healing, with or without wound inoculation with Staphylococcus aureus and Pseudomonas aeruginosa, two major pathogens involved in cutaneous wound bacterial infections.ResultsAseptic excision in C57BL/6 mouse skin induced early expression of IL-1β, TNFα and Oncostatin M (OSM), without detectable expression of IL-22 and IL-17A/F. S. aureus and P. aeruginosa wound inoculation not only increased the expression of IL-1β and OSM, but also induced a strong cutaneous expression of IL-22, IL-17A and IL-17F, along with an increased number of infiltrating IL-17A and/or IL-22-producing γδ T cells. The same cytokine expression pattern was observed in infected human skin wounds. When compared to uninfected wounds, mouse skin infection delayed the wound healing process. Injection of IL-1α, TNFα, OSM, IL-22 and IL-17 together in the wound edges induced delayed wound healing similar to that induced by the bacterial infection. Wound healing experiments in infected Rag2KO mice (deficient in lymphocytes) showed a wound healing kinetic similar to uninfected Rag2KO mice or WT mice. Rag2KO infected-skin lesions expressed lower levels of IL-17 and IL-22 than WT, suggesting that the expression of these cytokines is mainly dependent on γδ T cells in this model. Wound healing was not delayed in infected IL-17R/IL-22KO, comparable to uninfected control mice. Injection of recombinant IL-22 and IL-17 in infected wound edges of Rag2KO mice re-establish the delayed kinetic of wound healing, as in infected WT mice.ConclusionThese results demonstrate the synergistic and specific effects of IL-22 and IL-17 induced by bacterial infection delay the wound healing process, regardless of the presence of bacteria per se. Therefore, these cytokines play an unexpected role in delayed skin wound healing.
Collapse
Affiliation(s)
- Jean-Claude Lecron
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Laboratoire Immunologie et Inflammation, Centre Hospitalier et Universitaire (CHU) de Poitiers, Poitiers, France
- *Correspondence: Jean-Claude Lecron,
| | - Sandrine Charreau
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Qima-Bioalternatives (Qima Life Sciences), Gençay, France
| | - Jean-François Jégou
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| | - Nadjet Salhi
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| | - Isabelle Petit-Paris
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| | - Emmanuel Guignouard
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| | - Christophe Burucoa
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Laboratoire de Bactériologie, Centre Hospitalier et Universitaire (CHU) de Poitiers, Poitiers, France
| | - Laure Favot-Laforge
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| | - Charles Bodet
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| | - Anne Barra
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Laboratoire Immunologie et Inflammation, Centre Hospitalier et Universitaire (CHU) de Poitiers, Poitiers, France
| | - Vincent Huguier
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Service de Chirurgie Plastique, Centre Hospitalier et Universitaire (CHU) de Poitiers, Poitiers, France
| | - Jiad Mcheik
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Service de Chirurgie Pédiatrique, Centre Hospitalier et Universitaire CHU) de Poitiers, Poitiers, France
| | - Laure Dumoutier
- De Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Julien Garnier
- Qima-Bioalternatives (Qima Life Sciences), Gençay, France
| | - François-Xavier Bernard
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
- Qima-Bioalternatives (Qima Life Sciences), Gençay, France
| | - Bernhard Ryffel
- Laboratoire d'Immunologie et Neurogénétique Expérimentales et Moléculaire (INEM) - Unité Mixte de Recherche (UMR) 7355, Centre National de la Recherche Scientifique (CNRS) et Université d’Orléans, Orléans, France
| | - Franck Morel
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, UR15560, Université de Poitiers, Poitiers, France
| |
Collapse
|
47
|
Dong X, Limjunyawong N, Sypek EI, Wang G, Ortines RV, Youn C, Alphonse MP, Dikeman D, Wang Y, Lay M, Kothari R, Vasavda C, Pundir P, Goff L, Miller LS, Lu W, Garza LA, Kim BS, Archer NK, Dong X. Keratinocyte-derived defensins activate neutrophil-specific receptors Mrgpra2a/b to prevent skin dysbiosis and bacterial infection. Immunity 2022; 55:1645-1662.e7. [PMID: 35882236 PMCID: PMC9474599 DOI: 10.1016/j.immuni.2022.06.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 04/19/2022] [Accepted: 06/24/2022] [Indexed: 11/20/2022]
Abstract
Healthy skin maintains a diverse microbiome and a potent immune system to fight off infections. Here, we discovered that the epithelial-cell-derived antimicrobial peptides defensins activated orphan G-protein-coupled receptors (GPCRs) Mrgpra2a/b on neutrophils. This signaling axis was required for effective neutrophil-mediated skin immunity and microbiome homeostasis. We generated mutant mouse lines lacking the entire Defensin (Def) gene cluster in keratinocytes or Mrgpra2a/b. Def and Mrgpra2 mutant animals both exhibited skin dysbiosis, with reduced microbial diversity and expansion of Staphylococcus species. Defensins and Mrgpra2 were critical for combating S. aureus infections and the formation of neutrophil abscesses, a hallmark of antibacterial immunity. Activation of Mrgpra2 by defensin triggered neutrophil release of IL-1β and CXCL2 which are vital for proper amplification and propagation of the antibacterial immune response. This study demonstrated the importance of epithelial-neutrophil signaling via the defensin-Mrgpra2 axis in maintaining healthy skin ecology and promoting antibacterial host defense.
Collapse
Affiliation(s)
- Xintong Dong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nathachit Limjunyawong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth I Sypek
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gaofeng Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Roger V Ortines
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christine Youn
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Martin P Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dustin Dikeman
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yu Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mark Lay
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ruchita Kothari
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chirag Vasavda
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Priyanka Pundir
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Loyal Goff
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wuyuan Lu
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Luis A Garza
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brian S Kim
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nathan K Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
48
|
TLR2, TLR4, and NLRP3 mediated the balance between host immune-driven resistance and tolerance in Staphylococcus aureus-infected mice. Microb Pathog 2022; 169:105671. [PMID: 35811022 DOI: 10.1016/j.micpath.2022.105671] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 06/15/2022] [Accepted: 07/05/2022] [Indexed: 01/02/2023]
Abstract
Staphylococcus aureus (S. aureus) is a gram-positive pathogen that can cause infectious diseases in mammals. S. aureus-induced host innate immune responses have a relationship with Toll-like receptor 2 (TLR2), TLR4, and Nod-like receptor pyrin domain-containing protein 3 (NLRP3). However, the detailed roles of TLR2, TLR4, and NLRP3 in regulating the host inflammatory response to S. aureus infection remain unclear. Our data indicated that the S. aureus-induced mortality was aggravated by deficiency of TLR2, TLR4, and NLRP3 in mice. In the subsequent experiment, we found that during S. aureus infection, the roles of TLR2, TLR4, and NLRP3 seemed to be different at multiple timepoints. The deficiency of TLR2, TLR4, or NLRP3 attenuated the expression of High-mobility group box protein 1 (HMGB1) and Hyaluronic acid-binding protein 2 (HABP2), which is accompanied by decreased proinflammatory cytokine (TNF-α), chemokine (RANTES), and anti-inflammatory cytokine (IL-10) production in lungs and serum at 3 h and 6 h post-infection. However, with S. aureus infection prolonged (24 h post-infection), the trend was diametrically opposite. The results showed that deficiency of TLR2, TLR4, or NLRP3 aggravated HABP2 and HMGB1 expression, which is accompanied by enhanced proinflammatory cytokine (TNF-α), chemokine (RANTES), and anti-inflammatory cytokine (IL-10) production in lungs and serum. These results were consistent with the data observed in S. aureus-infected bone marrow-derived macrophages (BMDMs). All these results suggested that during S. aureus infection, TLR2, TLR4, and NLRP3 has time-dependent effect in regulating the balance between immune-driven resistance and tolerance.
Collapse
|
49
|
Kumar A, Datta A, Kumar S. A photo-reversible, sensitive, and selective sensor for copper ions in an aqueous medium. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
50
|
Shen P, Lin H, Bao Y, Hong H, Wu Z. Synthesis and immunological study of a glycosylated wall teichoic acid-based vaccine against Staphylococcus aureus. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|