1
|
Xu Y, Da X, Jian Y, Zhou W, Wu A, Wu Y, Peng Y, Liu X, Shi Y, Wang X, Zhou Q. A highly positively charged Ru(II) complex with photo-labile ligands for selective and efficient photo-inactivation of intracellular Staphylococcus aureus. J Inorg Biochem 2025; 268:112908. [PMID: 40209460 DOI: 10.1016/j.jinorgbio.2025.112908] [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: 12/14/2024] [Revised: 03/12/2025] [Accepted: 04/01/2025] [Indexed: 04/12/2025]
Abstract
Due to the protection afforded by host cells, intracellular Staphylococcus aureus (S. aureus), particularly methicillin-resistant S. aureus (MRSA), poses a significantly greater challenge to eliminate compared to the extracellular counterparts. It is highly desirable to develop novel antibacterial agents which are capable of selectively and efficiently eradicating intracellular bacteria, including drug-resistant strains, while being less prone to induce bacterial resistance. In this work, two Ru(II) complexes (Ru1 and Ru2) with photo-labile ligands were designed and synthesized. Both Ru1 and Ru2 could covalently bind to DNA after photo-induced ligand dissociation. Compared to Ru1, the incorporation of a triphenylamine group adorned with two positively charged cationic pyridine units significantly boosts the DNA binding constant, bacterial binding/uptake level, and subsequently, the antibacterial activity of Ru2. Ru2 could selectively photo-inactivate intracellular S. aureus and MRSA, being more efficient than vancomycin both in vitro and in vivo. Interestingly, after 20 days' treatment at sublethal concentrations, S. aureus cells exhibited no obvious drug resistance towards Ru2 upon irradiation. Such appealing results may provide new sights for developing novel antibacterial agents against intractable intracellular pathogens and also prevalent drug resistance.
Collapse
Affiliation(s)
- Yunli Xu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuwen Da
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Yao Jian
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wanpeng Zhou
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Aifeng Wu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yao Wu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yatong Peng
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiulian Liu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Shi
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuesong Wang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Qianxiong Zhou
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
2
|
Li S, Huang Y, Sun Q, Li Y, Xie H, Fu Q. Caspase-1 is critical for mice in the defense against Streptococcus equi subsp. zooepidemicus infection by promoting macrophage phagocytosis. Microb Pathog 2025; 203:107499. [PMID: 40122410 DOI: 10.1016/j.micpath.2025.107499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Streptococcus equi subsp. zooepidemicus (SEZ) is an important pathogen which is responsible for a wide range of diseases in various species. Macrophages are professional phagocytes that can engulf microorganisms and trigger responses leading to microbial death. Caspase-1 is considered as a proinflammatory factor that mediates antibacterial response to protect hosts from bacteria. Here, we revealed a novel role of Caspase-1 in mice against SEZ. Through both in vitro and in vivo infection assays, we demonstrated that the maturation and secretion of the cytokine IL-1β are critically dependent on Caspase-1 activation. The Caspase-1 deficient mice displayed attenuation of bactericidal activity against SEZ, mainly by decreasing the accumulation of macrophage. In addition to the recruitment of macrophages, deficiency of Caspase-1 also impaired the phagocytosis of SEZ by macrophages. Our study demonstrated that Caspase-1 is critical for mice to defense against SEZ depending on the recruitment and phagocytosis of macrophage.
Collapse
Affiliation(s)
- Shun Li
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China
| | - Yunfei Huang
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China
| | - Qinqin Sun
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China
| | - Yajuan Li
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China
| | - Honglin Xie
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China.
| | - Qiang Fu
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China.
| |
Collapse
|
3
|
Graham CT, Gordon S, Kubes P. A historical perspective of Kupffer cells in the context of infection. Cell Tissue Res 2025; 400:121-136. [PMID: 39392500 DOI: 10.1007/s00441-024-03924-4] [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: 06/28/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024]
Abstract
The Kupffer cell was first discovered by Karl Wilhelm von Kupffer in 1876, labeling them as "Sternzellen." Since their discovery as the primary macrophages of the liver, researchers have gradually gained an in-depth understanding of the identity, functions, and influential role of Kupffer cells, particularly in infection. It is becoming clear that Kupffer cells perform important tissue-specific functions in homeostasis and disease. Stationary in the sinusoids of the liver, Kupffer cells have a high phagocytic capacity and are adept in clearing the bloodstream of foreign material, toxins, and pathogens. Thus, they are indispensable to host defense and prevent the dissemination of bacteria during infections. To highlight the importance of this cell, this review will explore the history of the Kupffer cell in the context of infection beginning with its discovery to the present day.
Collapse
Affiliation(s)
- Carolyn T Graham
- Department of Physiology and Pharmacology, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada.
| | - Siamon Gordon
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, 259 Wenhua 1st Road Guishan Dist., Taoyuan, Taiwan
| | - Paul Kubes
- Department of Physiology and Pharmacology, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| |
Collapse
|
4
|
Douglas EJ, Palk N, Rudolph ER, Laabei M. Anti-staphylococcal fatty acids: mode of action, bacterial resistance and implications for therapeutic application. MICROBIOLOGY (READING, ENGLAND) 2025; 171:001563. [PMID: 40402078 PMCID: PMC12098983 DOI: 10.1099/mic.0.001563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/29/2025] [Indexed: 05/23/2025]
Abstract
Novel strategies to counter multidrug-resistant pathogens such as methicillin-resistant Staphylococcus aureus are urgently required. The antimicrobial properties of fatty acids (FAs) have long been recognized and offer significant promise as viable alternatives to, or potentiators of, conventional antibiotics. In this review, we examine the interplay between FAs and S. aureus, specifically detailing the underlying molecular mechanisms responsible for FA-mediated inhibition and the counteracting staphylococcal systems evolved to withstand FA onslaught. Finally, we present an update on the recent therapeutic FA applications to combat S. aureus infection, either as a monotherapy or in combination with antibiotics or host-derived antimicrobial peptides. Given the frequency of interaction between FAs and S. aureus during host colonization and infection, understanding FA mode of action and deciphering S. aureus FA resistance strategies are central in rationally designing future anti-staphylococcal FAs and FA-combination therapies.
Collapse
Affiliation(s)
- Edward J.A. Douglas
- Centre for Bacterial Resistance Biology, Imperial College London, London W2 1NY, UK
| | - Nathanael Palk
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Emily R. Rudolph
- Department of Life Sciences, University of Bath, Bath BA2 7AY, UK
| | - Maisem Laabei
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
5
|
Chow SH, Jeon Y, Deo P, Yeung ATY, Hale C, Sridhar S, Abraham G, Nickson J, Olivier FAB, Jiang JH, Ding Y, Han ML, Le Brun AP, Anderson D, Creek D, Tong J, Gabriel K, Li J, Traven A, Dougan G, Shen HH, Naderer T. Staphylococcal toxin PVL ruptures model membranes under acidic conditions through interactions with cardiolipin and phosphatidic acid. PLoS Biol 2025; 23:e3003080. [PMID: 40233125 PMCID: PMC12052211 DOI: 10.1371/journal.pbio.3003080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 05/05/2025] [Accepted: 02/24/2025] [Indexed: 04/17/2025] Open
Abstract
Panton-Valentine leukocidin (PVL) is a pore-forming toxin secreted by Staphylococcus aureus strains that cause severe infections. Bicomponent PVL kills phagocytes depending on cell surface receptors, such as complement 5a receptor 1 (C5aR1). How the PVL-receptor interaction enables assembly of the leukocidin complex, targeting of membranes, and insertion of a pore channel remains incompletely understood. Here, we demonstrate that PVL binds the anionic phospholipids, phosphatidic acid, and cardiolipin, under acidic conditions and targets lipid bilayers that mimic lysosomal and mitochondrial membranes, but not the plasma membrane. The PVL-lipid interaction was sufficient to enable leukocidin complex formation as determined by neutron reflectometry and the rupture of model membranes, independent of protein receptors. In phagocytes, PVL and its C5aR1 receptor were internalized depending on sphingomyelin and cholesterol, which were dispensable for the interaction of the toxin with the plasma membrane. Internalized PVL compromised the integrity of lysosomes and mitochondria before plasma membrane rupture. Preventing the acidification of organelles or the genetic loss of PVL impaired the escape of intracellular S. aureus from macrophages. Together, the findings advance our understanding of how an S. aureus toxin kills host cells and provide key insights into how leukocidins target membranes.
Collapse
Affiliation(s)
- Seong H. Chow
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Yusun Jeon
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Pankaj Deo
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Amy T. Y. Yeung
- The Wellcome Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
| | - Christine Hale
- The Wellcome Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
- Department of Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Sushmita Sridhar
- The Wellcome Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
- Department of Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Gilu Abraham
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Joshua Nickson
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Françios A. B. Olivier
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Jhih-Hang Jiang
- Department of Microbiology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Yue Ding
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Clayton, Australia
| | - Mei-Ling Han
- Department of Microbiology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Anton P. Le Brun
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organisation, Kirrawee DC, Australia
| | - Dovile Anderson
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Darren Creek
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Janette Tong
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Kip Gabriel
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Jian Li
- Centre to Impact AMR, Monash University, Clayton, Australia
- Department of Microbiology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Ana Traven
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| | - Gordon Dougan
- The Wellcome Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
- Department of Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Hsin-Hui Shen
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Clayton, Australia
| | - Thomas Naderer
- Department of Biochemistry and Molecular Biology, Infection Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Centre to Impact AMR, Monash University, Clayton, Australia
| |
Collapse
|
6
|
Rago F, Ahmed MY, Marinelli MA, Miller LM, Duray AM, Dresden BP, Constantinesco NJ, Sims PKF, Richwalls LJ, Kupul S, Kolls JK, Gopal R, Alcorn JF. CD209d/e are required for macrophage-mediated phagocytosis and activation during methicillin-resistant Staphylococcus aureus pulmonary host defense. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:703-713. [PMID: 40101753 PMCID: PMC12041778 DOI: 10.1093/jimmun/vkae061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 12/20/2024] [Indexed: 03/20/2025]
Abstract
Staphylococcus aureus is a commensal and opportunist pathogen of the upper respiratory tract. The recognition of pathogen-associated molecular patterns through pattern-recognition receptors is crucial for eliminating microorganisms such as S. aureus. DC-SIGN (CD209) is a pattern-recognition receptor that binds to a broad range of pathogens, promoting phagocytosis. Here we aimed to study the role of mouse homologues of DC-SIGN, CD209d/e, in a methicillin-resistant S. aureus (MRSA) pulmonary infection model. CD209d/e-/- and wild-type C57BL/6 mice were infected with MRSA and inflammatory parameters were evaluated. CD209d/e-/- mice had delayed bacterial burden and mortality together with increased frequency of neutrophils and decreased dendritic cells in the lung compared with control mice. iNOS+ macrophages, and regulatory T cell frequency were decreased in the lungs of CD209d/e-/- mice. CD209d/e-/- mice had increased levels of inflammatory cytokines in the lungs, but levels of IL-12p40 were decreased. MRSA reduced expression of interferon-γ and pattern-recognition receptors in CD209d/e-/- mice. MRSA uptake by phagocytes was decreased in the lungs of CD209d/e-/- versus control mice. CD209d/e-/- bone marrow derived macrophages showed impaired MRSA uptake and killing. These data suggest that CD209d/e are essential receptors to control inflammation by activating macrophages leading to MRSA uptake and killing.
Collapse
Affiliation(s)
- Flavia Rago
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Mohamed Y Ahmed
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Michael A Marinelli
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Leigh M Miller
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
- Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Alexis M Duray
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
- Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Brooke P Dresden
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
- Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Nicholas J Constantinesco
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Peyton K F Sims
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Lacee J Richwalls
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Saran Kupul
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Jay K Kolls
- Center for Translational Research in Infection & Inflammation, Tulane University School of Medicine, New Orleans, LA, United States
| | - Radha Gopal
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - John F Alcorn
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
7
|
An H, Huang Y, Zhao Z, Li K, Meng J, Huang X, Tian X, Zhou H, Wu J, Dai Q, Zhang JR. Splenic red pulp macrophages eliminate the liver-resistant Streptococcus pneumoniae from the blood circulation of mice. SCIENCE ADVANCES 2025; 11:eadq6399. [PMID: 40073120 PMCID: PMC11900858 DOI: 10.1126/sciadv.adq6399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025]
Abstract
Invasive infections by encapsulated bacteria are the major cause of human morbidity and mortality. The liver resident macrophages, Kupffer cells, form the hepatic firewall to clear many encapsulated bacteria in the blood circulation but fail to control certain high-virulence capsule types. Here we report that the spleen is the backup immune organ to clear the liver-resistant serotypes of Streptococcus pneumoniae (pneumococcus), a leading human pathogen. Asplenic mice failed to control the growth of the liver-resistant pneumococci in the blood circulation. Immunologic and genetic analyses identified splenic red pulp (RP) macrophages as the major phagocytes for bacterial clearance. Furthermore, the plasma natural antibodies against the cell wall phosphocholine and the complement system were necessary for RP macrophage-mediated immunity. These findings have provided a conceptual framework for the innate defense against blood bacterial infections, a mechanistic explanation for the hyper-susceptibility of asplenic individuals to S. pneumoniae, and a proof of concept for developing vaccines and therapeutic antibodies against encapsulated pathogens.
Collapse
Affiliation(s)
- Haoran An
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China
- Department of Microbiology and Infectious Disease Center, Peking University Health Science Center, Beijing 100191, China
- Center for Infectious Biology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Yijia Huang
- Department of Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhifeng Zhao
- Department of Automation, Tsinghua University, Beijing 100084, China
| | - Kunpeng Li
- Center for Infectious Biology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Jingjing Meng
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China
| | - Xueting Huang
- Center for Infectious Biology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Xianbin Tian
- Center for Infectious Biology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Hongyu Zhou
- Department of Automation, Tsinghua University, Beijing 100084, China
| | - Jiamin Wu
- Department of Automation, Tsinghua University, Beijing 100084, China
| | - Qionghai Dai
- Department of Automation, Tsinghua University, Beijing 100084, China
| | - Jing-Ren Zhang
- Center for Infectious Biology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
8
|
Rasquel-Oliveira FS, Ribeiro JM, Martelossi-Cebinelli G, Costa FB, Nakazato G, Casagrande R, Verri WA. Staphylococcus aureus in Inflammation and Pain: Update on Pathologic Mechanisms. Pathogens 2025; 14:185. [PMID: 40005560 PMCID: PMC11858194 DOI: 10.3390/pathogens14020185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/23/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Staphylococcus aureus (S. aureus) is a Gram-positive bacterium of significant clinical importance, known for its versatility and ability to cause a wide array of infections, such as osteoarticular, pulmonary, cardiovascular, device-related, and hospital-acquired infections. This review describes the most recent evidence of the pathogenic potential of S. aureus, which is commonly part of the human microbiota but can lead to severe infections. The prevalence of pathogenic S. aureus in hospital and community settings contributes to substantial morbidity and mortality, particularly in individuals with compromised immune systems. The immunopathogenesis of S. aureus infections involves intricate interactions with the host immune and non-immune cells, characterized by various virulence factors that facilitate adherence, invasion, and evasion of the host's defenses. This review highlights the complexity of S. aureus infections, ranging from mild to life-threatening conditions, and underscores the growing public health concern posed by multidrug-resistant strains, including methicillin-resistant S. aureus (MRSA). This article aims to provide an updated perspective on S. aureus-related infections, highlighting the main diseases linked to this pathogen, how the different cell types, virulence factors, and signaling molecules are involved in the immunopathogenesis, and the future perspectives to overcome the current challenges to treat the affected individuals.
Collapse
Affiliation(s)
- Fernanda S. Rasquel-Oliveira
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Jhonatan Macedo Ribeiro
- Department of Microbiology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil (G.N.)
| | - Geovana Martelossi-Cebinelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Fernanda Barbosa Costa
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Gerson Nakazato
- Department of Microbiology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil (G.N.)
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Center of Health Science, Londrina State University, Londrina 86038-440, PR, Brazil
| | - Waldiceu A. Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| |
Collapse
|
9
|
Erdene E, Munkhjargal O, Batnasan G, Dorjbal E, Oidov B, Byambaa A. Evaluation of Liposome-Encapsulated Vancomycin Against Methicillin-Resistant Staphylococcus aureus. Biomedicines 2025; 13:378. [PMID: 40002791 PMCID: PMC11853440 DOI: 10.3390/biomedicines13020378] [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: 01/09/2025] [Revised: 01/30/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Methicillin-resistant Staphylococcus aureus (MRSA) is a global health concern due to its resistance to conventional antibiotics. This study evaluated the efficacy of liposome-encapsulated vancomycin against MRSA using phospholipids extracted from egg yolk. Liposomes were prepared via the freeze-thaw method, yielding vesicles with an average diameter of 157.01 ± 33.04 nm and a polydispersity index (PDI) of 0.0442, indicating uniformity and stability. Antibacterial activity was assessed using the microdilution method. Liposome-encapsulated vancomycin demonstrated complete bacterial growth inhibition (100%) against MRSA ATCC 2758 at dilutions of 101 and 102, compared to only 50% inhibition by free vancomycin at 101. At higher dilutions (103), liposome-encapsulated vancomycin maintained 70% inhibition, whereas free vancomycin was ineffective. In vivo studies using a murine wound infection model revealed that wounds treated with liposome-encapsulated vancomycin achieved superior healing, with complete tissue regeneration observed by day 14. Histological analysis showed reduced inflammation and enhanced tissue recovery in liposome-encapsulated vancomycin-treated groups, compared to fibrosis and persistent necrosis in free vancomycin-treated groups. By enabling sustained drug release and improved bioavailability, liposomal formulations minimized required dosages and systemic toxicity, reducing the risk of resistance development. This study highlights the clinical potential of liposome-encapsulated vancomycin as a scalable, cost-effective treatment for MRSA, particularly in resource-limited settings.
Collapse
Affiliation(s)
- Enkhtaivan Erdene
- Department of Microbiology, Infection Prevention and Control, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14200, Mongolia;
- Department of Biomedicine, Etugen University, Ulaanbaatar 14200, Mongolia
| | - Odonchimeg Munkhjargal
- Mongolian Academy of Science, Institute of Chemistry and Chemical Technology, Ulaanbaatar 14200, Mongolia
| | - Galindev Batnasan
- Experimental Animal Center, Institute of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14200, Mongolia
| | - Enkhjargal Dorjbal
- Department of Pharmaceutical Chemistry and Pharmacognosy, School of Pharmacy, Mongolian National University of Medical Sciences, Ulaanbaatar 14200, Mongolia
| | - Baatarkhuu Oidov
- Department of Infectious Diseases, School of Medicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14200, Mongolia
| | - Ariunsanaa Byambaa
- Department of Microbiology, Infection Prevention and Control, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar 14200, Mongolia;
| |
Collapse
|
10
|
Mapar M, Rydzak T, Hommes JW, Surewaard BGJ, Lewis IA. Diverse molecular mechanisms underpinning Staphylococcus aureus small colony variants. Trends Microbiol 2025; 33:223-232. [PMID: 39393939 DOI: 10.1016/j.tim.2024.09.007] [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: 05/01/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/13/2024]
Abstract
Small colony variants (SCVs) of Staphylococcus aureus are a relatively rare but clinically significant growth morphotype. Infections with SCVs are frequently difficult to treat, inherently antibiotic-resistant, and can lead to persistent infections. Despite a long history of research, the molecular underpinnings of this morphotype and their impact on the clinical trajectory of infections remain unclear. However, a growing body of literature indicates that SCVs are caused by a diverse range of molecular factors. These recent findings suggest that SCVs should be thought of as an ensemble collection of loosely related phenotypes, and not as a single phenomenon. This review describes the diverse mechanisms currently known to contribute to SCVs and proposes an ensemble model for conceptualizing this morphotype.
Collapse
Affiliation(s)
- Maryam Mapar
- Alberta Centre for Advanced Diagnostics, Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Thomas Rydzak
- Alberta Centre for Advanced Diagnostics, Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Josefien W Hommes
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Bas G J Surewaard
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ian A Lewis
- Alberta Centre for Advanced Diagnostics, Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
11
|
Schmidt C, Harit K, Traidl S, Naumann M, Werfel T, Roesner LM, Nishanth G, Schlüter D. Ablation of the deubiquitinating enzyme cylindromatosis (CYLD) augments STAT1-mediated M1 macrophage polarization and fosters Staphylococcus aureus control. Front Immunol 2025; 16:1507989. [PMID: 39958342 PMCID: PMC11827430 DOI: 10.3389/fimmu.2025.1507989] [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: 10/08/2024] [Accepted: 01/07/2025] [Indexed: 02/18/2025] Open
Abstract
In atopic dermatitis (AD), lesional skin is frequently colonized by Staphylococcus aureus, which promotes clinical symptoms of the disease. The inflammatory milieu in the skin is characterized by a Th2 response, including M2 macrophages, which cannot eradicate S. aureus. Therefore, repolarization of macrophages toward the M1 phenotype may foster control of S. aureus. Our data show that the deubiquitinating enzyme cylindromatosis (CYLD) is strongly expressed in macrophages of AD patients and prevents the clearance of S. aureus. Mechanistically, CYLD impaired M1 macrophage polarization by K63-specific deubiquitination of STAT1 and activation of the NF-κB pathway via its interaction with TRAF6, NEMO, and RIPK2. Inhibition of STAT1 and NF-κB, independently, abolished the differences between S. aureus-infected CYLD-deficient and CYLD-competent M1 macrophages. Infection of Cyld-deficient and wild-type mice with S. aureus confirmed the protective CYLD function. Collectively, our study shows that CYLD impairs the control of S. aureus in macrophages of AD patients, identifying CYLD as a potential therapeutic target.
Collapse
Affiliation(s)
- Christina Schmidt
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Kunjan Harit
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Stephan Traidl
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Thomas Werfel
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Lennart M. Roesner
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Gopala Nishanth
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Dirk Schlüter
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
12
|
Yang X, Tang X, Yi S, Guo T, Liao Y, Wang Y, Zhang X. Maltodextrin-derived nanoparticles resensitize intracellular dormant Staphylococcus aureus to rifampicin. Carbohydr Polym 2025; 348:122843. [PMID: 39562116 DOI: 10.1016/j.carbpol.2024.122843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/01/2024] [Accepted: 10/06/2024] [Indexed: 11/21/2024]
Abstract
Intracellular bacteria are recognized as a crucial factor in the persistence and recurrence of infections. The efficacy of current antibiotic treatments faces substantial challenges due to the dormant state formation of intracellular bacteria. In this study, we devised a strategy aimed at reverting intracellular dormant bacteria to a metabolically active state, thereby increasing their vulnerability to antibiotics. We found that oligosaccharides, especially maltodextrin (MD), can be absorbed by dormant S. aureus, leading to their revival and restoration of sensitivity to rifampicin (Rif). We then synthesized a reactive oxygen species (ROS)-responsive MD-prodrug by covalently binding MD with 4-(hydroxymethyl) phenylboronic acid pinacol ester (MD-PBAP) and prepared a ROS-responsive nanoparticles (MDNP) using a nanoprecipitation and self-assembly method. Once internalized by host cells, MDNP was degraded to MD, reactivating dormant S. aureus, and enhancing their susceptibility to Rif. More importantly, MDNP treatment restored the sensitivity of intracellular persistent S. aureus to Rif in both a reservoir transfer model and whole-body infection model. Additionally, MDNP have demonstrated excellent biocompatibility in both in vitro and in vivo settings. These results offer a promising therapeutic avenue for managing persistent intracellular bacterial infections by reviving and resensitizing intracellular dormant bacteria to conventional antibiotics.
Collapse
Affiliation(s)
- Xiaodi Yang
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China
| | - Xiyu Tang
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China
| | - Sisi Yi
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China
| | - Tao Guo
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China
| | - Yue Liao
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China
| | - Yan Wang
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China
| | - Xiangjun Zhang
- College of Pharmacy, Chongqing Medical University, 400016 Chongqing, China.
| |
Collapse
|
13
|
Szymczak K, Rychłowski M, Zhang L, Nakonieczna J. Harnessing light-activated gallium porphyrins to combat intracellular Staphylococcus aureus using an in vitro keratinocyte infection model. Sci Rep 2025; 15:1295. [PMID: 39779728 PMCID: PMC11711192 DOI: 10.1038/s41598-024-84312-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
Staphylococcus aureus (S. aureus) can survive inside nonprofessional phagocytes such as keratinocytes, enabling it to evade antibiotics and cause recurrent infections once treatment stops. New antibacterial strategies to eliminate intracellular, multidrug-resistant bacteria are needed. This study used a keratinocyte model infected with methicillin-resistant S. aureus (MRSA) to test light-activated compounds, specifically heme-mimetic gallium (III) porphyrin (Ga3+CHP) and visible light, known as antimicrobial photodynamic inactivation (aPDI), for eliminating intracellular MRSA. Ga3+CHP was found to accumulate more in infected cells, particularly within lysosomal structures where MRSA resides. Flow cytometry and fluorescence microscopy revealed significant colocalization of MRSA and Ga3+CHP. Under aPDI, MRSA showed reduced adhesion to host cells and a 70% reduction in the GFP signal from intracellular bacteria. Additionally, light-activated Ga3+CHP significantly decreased the number of extracellular bacteria, reducing the potential for further infection. This study is the first to analyze aPDI toxicity in real time within an infection model, demonstrating that this method is neither cytotoxic nor phototoxic.
Collapse
Affiliation(s)
- Klaudia Szymczak
- Laboratory of Photobiology and Molecular Diagnostics, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdańsk, Poland
| | - Michał Rychłowski
- Laboratory of Virus Molecular Biology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdańsk, Poland
| | - Lei Zhang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), Tianjin University, Tianjin, China
| | - Joanna Nakonieczna
- Laboratory of Photobiology and Molecular Diagnostics, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdańsk, Poland.
| |
Collapse
|
14
|
Grosso R, Nguyen V, Ahmed SK, Wong-Beringer A. Novel Epigallocatechin Gallate (EGCG) Analogs with Improved Biochemical Properties for Targeting Extracellular and Intracellular Staphylococcus aureus. Appl Microbiol 2024; 4:1568-1581. [DOI: 10.3390/applmicrobiol4040107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Staphylococcus aureus is a leading cause of bloodstream infection (SAB), with up to 30% mortality. Despite treatment with standard antibiotics, one in three patients develops a persistent infection, which portends a five-fold increase in the risk of death. Persistent SAB has been attributed in part to the inability of antistaphylococcal antibiotics to eradicate intracellular S. aureus surviving inside macrophages. (-)- Epigallocatechin gallate (EGCG) is a catechin found in green tea that has been widely studied for its broad biological activities, ranging from anticancer to antibacterial activity. However, EGCG is greatly limited by its poor drug-like properties in terms of stability, membrane permeability, and bioavailability. In this study, we established through a series of in vitro experiments that structural modifications of EGCG enhanced drug-like properties while maintaining or improving its antistaphylococcal activity. Our lead EGCG analogs (MCC-1 and MCC-2) showed improved biochemical properties along with increased potency against extracellular S. aureus and restored susceptibility of β-lactam agents to methicillin-resistant S. aureus (MRSA). Importantly, the lead analogs but not EGCG potentiated macrophage- and antibiotic-mediated clearance of intracellular bacteria. Overall, EGCG analogs showed promise for further development as adjunctive therapy candidates for the treatment of SAB.
Collapse
Affiliation(s)
- Riley Grosso
- Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern Los Angeles, Los Angeles, CA 90033, USA
| | - Vy Nguyen
- Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern Los Angeles, Los Angeles, CA 90033, USA
| | - Syed Kaleem Ahmed
- Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern Los Angeles, Los Angeles, CA 90033, USA
| | - Annie Wong-Beringer
- Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern Los Angeles, Los Angeles, CA 90033, USA
| |
Collapse
|
15
|
Yang J, Chen L, Cai Z, Pang L, Huang Y, Xiao P, Wang J, Huang W, Cui W, Hu N. Precise Clearance of Intracellular MRSA via Internally and Externally Mediated Bioorthogonal Activation of Micro/Nano Hydrogel Microspheres. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402370. [PMID: 39342650 PMCID: PMC11600240 DOI: 10.1002/advs.202402370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 08/25/2024] [Indexed: 10/01/2024]
Abstract
Traditional high-dose antibiotic treatments of intracellular methicillin-resistant staphylococcus aureus (MRSA) are highly inefficient and associated with a high rate of infection relapse. As an effective antibacterial technology, sonodynamic therapy (SDT) may be able to break the dilemma. However, indiscriminate reactive oxygen species (ROS) release leads to potential side effects. This study incorporates Staphylococcal Protein A antibody-modified Cu2+/tetracarboxyphenylporphyrin nanoparticles (Cu(II)NS-SPA) into hydrogel microspheres (HAMA@Cu(II)NS-SPA) to achieve precise eradication of intracellular bacteria. This eradication is under bioorthogonal activation mediated by bacillithiol (BSH) (internally) and ultrasound (US) (externally). To specify, the US responsiveness of Cu(II)NS-SPA is restored when it is reduced to Cu(I)NS-SPA by the BSH secreted characteristically by intracellular MRSA, thus forming a bioorthogonal activation with the external US, which confines ROS production within the infected MΦ. Under external US activation at 2 W cm-2, over 95% of intracellular MRSA can be cleared. In vivo, a single injection of HAMA@Cu(II)NS-SPA achieves up to two weeks of antibacterial sonodynamic therapy, reducing pro-inflammatory factor expression by 90%, and peri-implant bone trabeculae numbers exceed the control group by five times. In summary, these micro/nano hydrogel microspheres mediated by internal and external bioorthogonal activation can precisely eliminate intracellular MRSA, effectively treating multi-drug resistant intracellular bacterial infections.
Collapse
Affiliation(s)
- Jianye Yang
- Department of OrthopaedicsThe First Affiliated Hospital of Chongqing Medical UniversityOrthopedic Laboratory of Chongqing Medical UniversityChongqing400016P. R. China
| | - Li Chen
- Department of OrthopaedicsThe First Affiliated Hospital of Chongqing Medical UniversityOrthopedic Laboratory of Chongqing Medical UniversityChongqing400016P. R. China
| | - Zhengwei Cai
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Libin Pang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Yanran Huang
- Department of OrthopaedicsThe First Affiliated Hospital of Chongqing Medical UniversityOrthopedic Laboratory of Chongqing Medical UniversityChongqing400016P. R. China
| | - Pengcheng Xiao
- Department of OrthopaedicsThe First Affiliated Hospital of Chongqing Medical UniversityOrthopedic Laboratory of Chongqing Medical UniversityChongqing400016P. R. China
| | - Juan Wang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Wei Huang
- Department of OrthopaedicsThe First Affiliated Hospital of Chongqing Medical UniversityOrthopedic Laboratory of Chongqing Medical UniversityChongqing400016P. R. China
| | - Wenguo Cui
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Ning Hu
- Department of OrthopaedicsThe First Affiliated Hospital of Chongqing Medical UniversityOrthopedic Laboratory of Chongqing Medical UniversityChongqing400016P. R. China
| |
Collapse
|
16
|
Lu KY, Yang X, Eldridge MJG, Wagner NJ, Hardy B, Axtman M, Rowe SE, Wang X, Fowler VG, Helaine S, Pearce KH, Conlon BP. A host-directed adjuvant resuscitates and sensitizes intracellular bacterial persisters to antibiotics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.30.615828. [PMID: 39554024 PMCID: PMC11565766 DOI: 10.1101/2024.09.30.615828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
There are two major problems in the field of antimicrobial chemotherapy-antibiotic resistance and antibiotic tolerance. In the case of antibiotic tolerance, antibiotics fail to kill the bacteria as their phenotypic state affords them protection from the bactericidal activity of the antibiotic. Antibiotic tolerance can affect an entire bacterial population, or a subset of cells known as persister cells. Interaction with the host induces the formation of persister cells in numerous pathogens, with reactive oxygen and nitrogen species production being heavily implicated in the collapse of bacterial energy levels and entrance into an antibiotic tolerant state. Here, we developed a high-throughput screen to identify energy modulators for intracellular Staphylococcus aureus . The identified compound, KL1 , increases intracellular bacterial energy and sensitizes the persister population to antibiotics, without causing cytotoxicity or bacterial outgrowth. We demonstrate that KL1 exhibits adjuvant activity in a murine model of S. aureus bacteremia and intracellular infection of Salmonella Typhimurium . Transcriptomic analysis and further studies on its mechanism of action reveal that KL1 modulates host immune response genes and suppresses the production of reactive species in host macrophages, alleviating one of the major stressors that induce antibiotic tolerance. Our findings highlight the potential to target intracellular persister cells by stimulating their metabolism and encourage larger efforts to address antibiotic tolerance at the host-pathogen interface, particularly within the intracellular milieu.
Collapse
|
17
|
Dai F, Zhang X, Ma G, Li W. ACOD1 mediates Staphylococcus aureus-induced inflammatory response via the TLR4/NF-κB signaling pathway. Int Immunopharmacol 2024; 140:112924. [PMID: 39133958 DOI: 10.1016/j.intimp.2024.112924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/01/2024]
Abstract
Staphylococcus aureus (SA) is a common Gram-positive bacterium that activates inflammatory cells, expressing various cytokines and inducing an inflammatory response. Recent research revealed aconitate decarboxylase 1 (ACOD1) as a regulator of the immune response through various metabolic pathways, playing a dual role in the inflammatory response. However, the mechanism by which ACOD1 participates in the regulation of SA-induced inflammatory responses in macrophages remains unknown. Therefore, this study aims to investigate the function and underlying regulatory mechanisms of ACOD1 in SA-induced inflammatory response. This study reveals that SA induced a macrophage inflammatory response and upregulated ACOD1 expression. ACOD1 knockdown significantly inhibited SA-induced macrophage inflammatory response, attenuated SA-induced nuclear envelope wrinkling, and plasma membrane rupture, and suppressed the TLR4/NF-κB signaling pathway. Furthermore, ACOD1 knockdown reduced the inflammatory response and alleviated lung tissue injury and cellular damage, leading to decreased bacterial loads in the lungs of SA-infected mice. Collectively, these findings demonstrate that SA induces an inflammatory response in macrophages and increases ACOD1 expression. ACOD1 enhances SA-induced inflammatory responses via the TLR4/NF-κB signaling pathway. Our findings highlight the significant role of ACOD1 in mediating the inflammatory response in SA-infected macrophages and elucidate its molecular mechanism in regulating the SA-induced inflammatory response.
Collapse
Affiliation(s)
- Fan Dai
- School of Life Sciences, Ningxia University, Yinchuan, Ningxia 750021, China; Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Xuyang Zhang
- School of Life Sciences, Ningxia University, Yinchuan, Ningxia 750021, China; Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Guilan Ma
- School of Life Sciences, Ningxia University, Yinchuan, Ningxia 750021, China; Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, Ningxia 750021, China
| | - Wu Li
- School of Life Sciences, Ningxia University, Yinchuan, Ningxia 750021, China; Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, Ningxia University, Yinchuan, Ningxia 750021, China.
| |
Collapse
|
18
|
Beadell B, Yamauchi J, Wong-Beringer A. Comparative in vitro efficacy of antibiotics against the intracellular reservoir of Staphylococcus aureus. J Antimicrob Chemother 2024; 79:2471-2478. [PMID: 39073778 PMCID: PMC11441993 DOI: 10.1093/jac/dkae241] [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: 02/27/2024] [Accepted: 06/26/2024] [Indexed: 07/30/2024] Open
Abstract
Staphylococcus aureus (SA) is a leading cause of bloodstream infection. The liver represents the sentinel immune organ for clearance of bloodstream pathogens and eradication of intracellular SA from liver-resident macrophages (Kupffer cells, KCs) eliminates the likely pathogenic reservoir that contributes to persistent bacteraemia. OBJECTIVES We assessed antimicrobial activity at phagolysosome-mimicking pH, intracellular penetration, and SA eradication within KCs in vitro for clinically prescribed antistaphylococcal agents alone or in combination: vancomycin, daptomycin, ceftaroline, ceftobiprole, oritavancin, oxacillin, cefazolin; rifampin and fosfomycin. METHODS pH-adjusted broth microdilution assays, intracellular bioaccumulation assays, and intracellular killing assays against clinical bloodstream isolates were performed using a murine KC line with study agents. RESULTS Rifampin and β-lactams exhibited enhanced activity [2- to 16-fold minimum inhibitory concentrations (MIC) decrease] at phagolysosomal pH while vancomycin, oritavancin, daptomycin and fosfomycin demonstrated reduced activity (2- to 32-fold MIC increase in order of least to greatest potency reduction). All agents evaluated had poor to modest intracellular to extracellular concentration ratios (0.024-7.8), with exceptions of rifampin and oritavancin (intracellular to extracellular ratios of 17.4 and 78.2, respectively). Finally, we showed that the first-line treatment for SA bacteraemia (SAB), vancomycin, performed worse than all other tested antibiotics in eradicating intracellular SA at human Cmax concentration (0.20 log cfu decrease), while oritavancin performed better than all other agents alone (2.05 versus 1.06-1.36 log cfu decrease). CONCLUSIONS Our findings raise concerns about the efficacy of commonly prescribed antibiotics against intracellular SA reservoirs and emphasize the need to consider targeting pathogen eradication from the liver to achieve early control of SAB.
Collapse
Affiliation(s)
- Brent Beadell
- Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, USA
| | - Joe Yamauchi
- Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, USA
| | - Annie Wong-Beringer
- Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, USA
- Department of Pharmacy, Huntington Hospital, Pasadena, CA, USA
| |
Collapse
|
19
|
Wang P, Xie C, Zhang Y, Li H, Lu Y, Sun L, Hu X, Nie T, Li C, Li G, Lu X, Pang J, Yang X, Yu L, Li X, Wang X, You X. Negatively charged nanodiscs for the reduction of toxicity and enhanced efficacy of polymyxin B against Acinetobacter baumannii sepsis. Acta Biomater 2024; 184:323-334. [PMID: 38901753 DOI: 10.1016/j.actbio.2024.06.017] [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: 03/14/2024] [Revised: 05/27/2024] [Accepted: 06/12/2024] [Indexed: 06/22/2024]
Abstract
The treatment of sepsis caused by multidrug-resistant (MDR) Gram-negative bacterial infections remains challenging. With these pathogens exhibiting resistance to carbapenems and new generation cephalosporins, the traditional antibiotic polymyxin B (PMB) has reemerged as a critical treatment option. However, its severe neurotoxicity and nephrotoxicity greatly limit the clinical application. Therefore, we designed negatively charged high-density lipoprotein (HDL) mimicking nanodiscs as a PMB delivery system, which can simultaneously reduce toxicity and enhance drug efficacy. The negative charge prevented the PMB release in physiological conditions and binding to cell membranes, significantly reducing toxicity in mammalian cells and mice. Notably, nanodisc-PMB exhibits superior efficacy than free PMB in sepsis induced by carbapenem-resistant Acinetobacter baumannii (CRAB) strains. Nanodisc-PMB shows promise as a treatment for carbapenem-resistant Gram-negative bacterial sepsis, especially caused by Acinetobacter baumannii, and the nanodiscs could be repurposed for other toxic antibiotics as an innovative delivery system. STATEMENT OF SIGNIFICANCE: Multidrug-resistant Gram-negative bacteria, notably carbapenem-resistant Acinetobacter baumannii, currently pose a substantial challenge due to the scarcity of effective treatments, rendering Polymyxins a last-resort antibiotic option. However, their therapeutic application is significantly limited by severe neurotoxic and nephrotoxic side effects. Prevailing polymyxin delivery systems focus on either reducing toxicity or enhancing bioavailability yet fail to simultaneously achieve both. In this scenario, we have developed a distinctive HDL-mimicking nanodisc for polymyxin B, which not only significantly reduces toxicity but also improves efficacy against Gram-negative bacteria, especially in sepsis caused by CRAB. This research offers an innovative drug delivery system for polymyxin B. Such advancement could notably improve the therapeutic landscape and make a significant contribution to the arsenal against these notorious pathogens.
Collapse
Affiliation(s)
- Penghe Wang
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Chunyang Xie
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Youwen Zhang
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Haibin Li
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yun Lu
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Lang Sun
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xinxin Hu
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Tongying Nie
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Congran Li
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Guoqing Li
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xi Lu
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jing Pang
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xinyi Yang
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Liyan Yu
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xue Li
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Xiukun Wang
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Xuefu You
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Division for Medicinal Microorganisms Related Strains, CAMS Collection Center of Pathogenic Microorganisms, Beijing 100050, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
20
|
Lacey KA, Pickrum AM, Gonzalez S, Bartnicki E, Castellaw AH, Rodrick TC, Jones DR, Khanna KM, Torres VJ. Dietary and water restriction leads to increased susceptibility to antimicrobial resistant pathogens. SCIENCE ADVANCES 2024; 10:eadi7438. [PMID: 39047095 PMCID: PMC11268424 DOI: 10.1126/sciadv.adi7438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 06/18/2024] [Indexed: 07/27/2024]
Abstract
Dehydration and malnutrition are common and often underdiagnosed in hospital settings. Multidrug-resistant bacterial infections result in more than 35,000 deaths a year in nosocomial patients. The effect of temporal dietary and water restriction (DWR) on susceptibility to multidrug-resistant pathogens is unknown. We report that DWR markedly increased susceptibility to systemic infection by ESKAPE pathogens. Using a murine bloodstream model of methicillin-resistant Staphylococcus aureus infection, we show that DWR leads to significantly increased mortality and morbidity. DWR causes increased bacterial burden, severe pathology, and increased numbers of phagocytes in the kidney. DWR appears to alter the functionality of these phagocytes and is therefore unable to control infection. Mechanistically, we show that DWR impairs the ability of macrophages to phagocytose multiple bacterial pathogens and efferocytose apoptotic neutrophils. Together, this work highlights the crucial impact that diet and hydration play in protecting against infection.
Collapse
Affiliation(s)
- Keenan A. Lacey
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Adam M. Pickrum
- Department of Host-Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Sandra Gonzalez
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Eric Bartnicki
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ashley H. Castellaw
- Department of Host-Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Tori C. Rodrick
- Metabolomics Core Resource Laboratory, New York University Langone Health, New York, NY 10016, USA
| | - Drew R. Jones
- Metabolomics Core Resource Laboratory, New York University Langone Health, New York, NY 10016, USA
| | - Kamal M. Khanna
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Department of Host-Microbe Interactions, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
21
|
Wang Y, Heymann F, Peiseler M. Intravital imaging: dynamic insights into liver immunity in health and disease. Gut 2024; 73:1364-1375. [PMID: 38777574 DOI: 10.1136/gutjnl-2023-331739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
Inflammation is a critical component of most acute and chronic liver diseases. The liver is a unique immunological organ with a dense vascular network, leading to intense crosstalk between tissue-resident immune cells, passenger leucocytes and parenchymal cells. During acute and chronic liver diseases, the multifaceted immune response is involved in disease promoting and repair mechanisms, while upholding core liver immune functions. In recent years, single-cell technologies have unravelled a previously unknown heterogeneity of immune cells, reshaping the complexity of the hepatic immune response. However, inflammation is a dynamic biological process, encompassing various immune cells, orchestrated in temporal and spatial dimensions, and driven by multiorgan signals. Intravital microscopy (IVM) has emerged as a powerful tool to investigate immunity by visualising the dynamic interplay between different immune cells and their surroundings within a near-natural environment. In this review, we summarise the experimental considerations to perform IVM and highlight recent technological developments. Furthermore, we outline the unique contributions of IVM to our understanding of liver immunity. Through the lens of liver disease, we discuss novel immune-mediated disease mechanisms uncovered by imaging-based studies.
Collapse
Affiliation(s)
- Yuting Wang
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Felix Heymann
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Moritz Peiseler
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité, Berlin, Germany
| |
Collapse
|
22
|
Zhu J, Xie R, Gao R, Zhao Y, Yodsanit N, Zhu M, Burger JC, Ye M, Tong Y, Gong S. Multimodal nanoimmunotherapy engages neutrophils to eliminate Staphylococcus aureus infections. NATURE NANOTECHNOLOGY 2024; 19:1032-1043. [PMID: 38632494 DOI: 10.1038/s41565-024-01648-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 03/12/2024] [Indexed: 04/19/2024]
Abstract
The increasing prevalence of antimicrobial resistance in Staphylococcus aureus necessitates alternative therapeutic approaches. Neutrophils play a crucial role in the fight against S. aureus but suffer from deficiencies in function leading to increased infection. Here we report a nanoparticle-mediated immunotherapy aimed at potentiating neutrophils to eliminate S. aureus. The nanoparticles consist of naftifine, haemoglobin (Hb) and a red blood cell membrane coating. Naftifine disrupts staphyloxanthin biosynthesis, Hb reduces bacterial hydrogen sulfide levels and the red blood cell membrane modifies bacterial lipid composition. Collectively, the nanoparticles can sensitize S. aureus to host oxidant killing. Furthermore, in the infectious microenvironment, Hb triggers lipid peroxidation in S. aureus, promoting neutrophil chemotaxis. Oxygen supplied by Hb can also significantly enhance the bactericidal capability of the recruited neutrophils by restoring neutrophil respiratory burst via hypoxia relief. This multimodal nanoimmunotherapy demonstrates excellent therapeutic efficacy in treating antimicrobial-resistant S. aureus persisters, biofilms and S. aureus-induced infection in mice.
Collapse
Affiliation(s)
- Jingcheng Zhu
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Ruosen Xie
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ruixuan Gao
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Yi Zhao
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Nisakorn Yodsanit
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Min Zhu
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Jacobus C Burger
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Mingzhou Ye
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Yao Tong
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Shaoqin Gong
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA.
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA.
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, USA.
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
23
|
Helaine S, Conlon BP, Davis KM, Russell DG. Host stress drives tolerance and persistence: The bane of anti-microbial therapeutics. Cell Host Microbe 2024; 32:852-862. [PMID: 38870901 PMCID: PMC11446042 DOI: 10.1016/j.chom.2024.04.019] [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: 03/03/2024] [Revised: 04/03/2024] [Accepted: 04/25/2024] [Indexed: 06/15/2024]
Abstract
Antibiotic resistance, typically associated with genetic changes within a bacterial population, is a frequent contributor to antibiotic treatment failures. Antibiotic persistence and tolerance, which we collectively term recalcitrance, represent transient phenotypic changes in the bacterial population that prolong survival in the presence of typically lethal concentrations of antibiotics. Antibiotic recalcitrance is challenging to detect and investigate-traditionally studied under in vitro conditions, our understanding during infection and its contribution to antibiotic failure is limited. Recently, significant progress has been made in the study of antibiotic-recalcitrant populations in pathogenic species, including Mycobacterium tuberculosis, Staphylococcus aureus, Salmonella enterica, and Yersiniae, in the context of the host environment. Despite the diversity of these pathogens and infection models, shared signals and responses promote recalcitrance, and common features and vulnerabilities of persisters and tolerant bacteria have emerged. These will be discussed here, along with progress toward developing therapeutic interventions to better treat recalcitrant pathogens.
Collapse
Affiliation(s)
- Sophie Helaine
- Department of Microbiology, Harvard Medical School, Boston, MA, USA.
| | - Brian P Conlon
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.
| | - Kimberly M Davis
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | - David G Russell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
24
|
Pormohammad A, Moradi M, Hommes JW, Pujol E, Naesens L, Vázquez S, Surewaard BGJ, Zarei M, Vazquez-Carrera M, Turner RJ. Novel pentafluorosulfanyl-containing triclocarban analogs selectively kill Gram-positive bacteria. Microbiol Spectr 2024; 12:e0007124. [PMID: 38700321 PMCID: PMC11237694 DOI: 10.1128/spectrum.00071-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 03/28/2024] [Indexed: 05/05/2024] Open
Abstract
Novel antimicrobial agents are needed to combat antimicrobial resistance. This study tested novel pentafluorosulfanyl-containing triclocarban analogs for their potential antibacterial efficacy. Standard procedures were used to produce pentafluorosulfanyl-containing triclocarban analogs. Twenty new compounds were tested against seven Gram-positive and Gram-negative indicator strains as well as 10 clinical isolates for their antibacterial and antibiofilm activity. Mechanistic investigations focused on damage to cell membrane, oxidizing reduced thiols, iron-sulfur clusters, and oxidative stress to explain the compounds' activity. Safety profiles were assessed using cytotoxicity experiments in eukaryotic cell lines. Following screening, selected components had significantly better antibacterial and antibiofilm activity against Gram-positive bacteria in lower concentrations in comparison to ciprofloxacin and gentamycin. For instance, one compound had a minimum inhibitory concentration of <0.0003 mM, but ciprofloxacin had 0.08 mM. Mechanistic studies show that these novel compounds do not affect reduced thiol content, iron-sulfur clusters, or hydrogen peroxide pathways. Their impact comes from Gram-positive bacterial cell membrane damage. Tests on cell culture toxicity and host component safety showed promise. Novel diarylurea compounds show promise as Gram-positive antimicrobials. These compounds offer prospects for study and optimization. IMPORTANCE The rise of antibiotic resistance among bacterial pathogens poses a significant threat to global health, underscoring the urgent need for novel antimicrobial agents. This study presents research on a promising class of novel compounds with potent antibacterial properties against Gram-positive bacteria, notably Staphylococcus aureus and MRSA. What sets these novel analogs apart is their superior efficacy at substantially lower concentrations compared with commonly used antibiotics like ciprofloxacin and gentamycin. Importantly, these compounds act by disrupting the bacterial cell membrane, offering a unique mechanism that could potentially circumvent existing resistance mechanisms. Preliminary safety assessments also highlight their potential for therapeutic use. This study not only opens new avenues for combating antibiotic-resistant infections but also underscores the importance of innovative chemical approaches in addressing the global antimicrobial resistance crisis.
Collapse
Affiliation(s)
- Ali Pormohammad
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
- MHCombiotic Inc., Calgary, Alberta, Canada
| | - Melika Moradi
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Science, Ahvaz, Iran
| | - Josefien W. Hommes
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Eugènia Pujol
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
| | - Lieve Naesens
- Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Santiago Vázquez
- Laboratori de Química Farmacèutica (Unitat Associada al CSIC), Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
| | - Bas G. J. Surewaard
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Mohammad Zarei
- Renal Division, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- John B. Little Center for Radiation Sciences, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Manuel Vazquez-Carrera
- Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
- Pharmacology Unit, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
- Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
| | - Raymond J. Turner
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
25
|
Tuffs SW, Dufresne K, Rishi A, Walton NR, McCormick JK. Novel insights into the immune response to bacterial T cell superantigens. Nat Rev Immunol 2024; 24:417-434. [PMID: 38225276 DOI: 10.1038/s41577-023-00979-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2023] [Indexed: 01/17/2024]
Abstract
Bacterial T cell superantigens (SAgs) are a family of microbial exotoxins that function to activate large numbers of T cells simultaneously. SAgs activate T cells by direct binding and crosslinking of the lateral regions of MHC class II molecules on antigen-presenting cells with T cell receptors (TCRs) on T cells; these interactions alter the normal TCR-peptide-MHC class II architecture to activate T cells in a manner that is independent of the antigen specificity of the TCR. SAgs have well-recognized, central roles in human diseases such as toxic shock syndrome and scarlet fever through their quantitative effects on the T cell response; in addition, numerous other consequences of SAg-driven T cell activation are now being recognized, including direct roles in the pathogenesis of endocarditis, bloodstream infections, skin disease and pharyngitis. In this Review, we summarize the expanding family of bacterial SAgs and how these toxins can engage highly diverse adaptive immune receptors. We highlight recent findings regarding how SAg-driven manipulation of the adaptive immune response may operate in multiple human diseases, as well as contributing to the biology and life cycle of SAg-producing bacterial pathogens.
Collapse
Affiliation(s)
- Stephen W Tuffs
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Karine Dufresne
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Aanchal Rishi
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Nicholas R Walton
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - John K McCormick
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
26
|
Wang J, Jiang H, Chen Y, Zhu X, Wu Q, Chen W, Zhao Q, Wang J, Qin P. CRISPR/Cas9-mediated SERS/colorimetric dual-mode lateral flow platform combined with smartphone for rapid and sensitive detection of Staphylococcus aureus. Biosens Bioelectron 2024; 249:116046. [PMID: 38241798 DOI: 10.1016/j.bios.2024.116046] [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: 11/26/2023] [Revised: 01/09/2024] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
Pathogenic bacteria infections pose a significant threat to global public health, making the development of rapid and reliable detection methods urgent. Here, we developed a surface-enhanced Raman scattering (SERS) and colorimetric dual-mode platform, termed smartphone-integrated CRISPR/Cas9-mediated lateral flow strip (SCC-LFS), and applied it to the ultrasensitive detection of Staphylococcus aureus (S. aureus). Strategically, functionalized silver-coated gold nanostar (AuNS@Ag) was prepared and used as the labeling material for LFS assay. In the presence of S. aureus, target gene-induced amplicons can be accurately recognized and unwound by the user-defined CRISPR/Cas9 system, forming intermediate bridges that bind many AuNS@Ag to the test line (T-line) of the strip. As a result, the T-line was colored and a recognizable SERS signal was obtained using a smartphone-integrated portable Raman spectrometer. This design not only maintains the simplicity of visual readout, but also integrates the quantitative capability of SERS, enabling the user to flexibly select the assay mode as needed. With this method, S. aureus down to 1 CFU/mL can be detected by both colorimetric and SERS modes, which is better than most existing methods. By incorporating a rapid extraction procedure, the entire assay can be completed in 45 min. The robustness and practicality of the method were further demonstrated by various real samples, indicating its considerable potential toward reliable screening of S. aureus.
Collapse
Affiliation(s)
- Junfeng Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, PR China; School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui, PR China
| | - Han Jiang
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, PR China
| | - Yuhong Chen
- School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui, PR China
| | - Xiaofan Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, PR China
| | - Qian Wu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, Anhui, PR China
| | - Wei Chen
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, Anhui, PR China.
| | - Qihong Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, PR China
| | - Jie Wang
- School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui, PR China.
| | - Panzhu Qin
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, PR China.
| |
Collapse
|
27
|
Bayer J, Becker J, Liu X, Gritsch L, Daiber E, Korn N, Oesterhelt F, Fraunholz M, Weber A, Wolz C. Differential survival of Staphylococcal species in macrophages. Mol Microbiol 2024; 121:470-480. [PMID: 37898563 DOI: 10.1111/mmi.15184] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/05/2023] [Accepted: 10/02/2023] [Indexed: 10/30/2023]
Abstract
Staphylococcus aureus is considered an extracellular pathogen, yet the bacterium is able to survive within and escape from host cells. An agr/sae mutant of strain USA300 is unable to escape from macrophages but can replicate and survive within. We questioned whether such "non-toxic" S. aureus resembles the less pathogenic coagulase-negative Staphylococcal (CoNS) species like S. epidermidis, S. carnosus, S. lugdunensis, S. capitis, S. warneri, or S. pettenkoferi. We show that the CoNS are more efficiently killed in macrophage-like THP-1 cells or in human primary macrophages. Mutations in katA, copL, the regulatory system graRS, or sigB did not impact bacterial survival in THP-1 cells. Deletion of the superoxide dismutases impaired S. aureus survival in primary macrophages but not in THP-1 cells. However, expression of the S. aureus-specific sodM in S. epidermidis was not sufficient to protect this species from being killed. Thus, at least in those cells, better bacterial survival of S. aureus could not be linked to higher protection from ROS. However, "non-toxic" S. aureus was found to be insensitive to pH, whereas most CoNS were protected when phagosomal acidification was inhibited. Thus, species differences are at least partially linked to differences in sensitivity to acidification.
Collapse
Affiliation(s)
- Janina Bayer
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
| | - Janna Becker
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
| | - Xiao Liu
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Tübingen, Germany
| | - Lisa Gritsch
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
| | - Ellen Daiber
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
| | - Natalya Korn
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
| | - Filipp Oesterhelt
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
| | - Martin Fraunholz
- Department of Microbiology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Alexander Weber
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
- Institute of Immunology, Department of Innate Immunity, University of Tübingen, Tübingen, Germany
| | - Christiane Wolz
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
| |
Collapse
|
28
|
Yamazaki Y, Ito T, Tamai M, Nakagawa S, Nakamura Y. The role of Staphylococcus aureus quorum sensing in cutaneous and systemic infections. Inflamm Regen 2024; 44:9. [PMID: 38429810 PMCID: PMC10905890 DOI: 10.1186/s41232-024-00323-8] [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: 12/18/2023] [Accepted: 02/15/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Staphylococcus aureus is a leading cause of human bacterial infections worldwide. It is the most common causative agent of skin and soft tissue infections, and can also cause various other infections, including pneumonia, osteomyelitis, as well as life-threatening infections, such as sepsis and infective endocarditis. The pathogen can also asymptomatically colonize human skin, nasal cavity, and the intestine. S. aureus colonizes approximately 20-30% of human nostrils, being an opportunistic pathogen for subsequent infection. Its strong ability to silently spread via human contact makes it difficult to eradicate S. aureus. A major concern with S. aureus is its capacity to develop antibiotic resistance and adapt to diverse environmental conditions. The variability in the accessory gene regulator (Agr) region of the genome contributes to a spectrum of phenotypes within the bacterial population, enhancing the likelihood of survival in different environments. Agr functions as a central quorum sensing (QS) system in S. aureus, allowing bacteria to adjust gene expression in response to population density. Depending on Agr expression, S. aureus secretes various toxins, contributing to virulence in infectious diseases. Paradoxically, expressing Agr may be disadvantageous in certain situations, such as in hospitals, causing S. aureus to generate Agr mutants responsible for infections in healthcare settings. MAIN BODY This review aims to demonstrate the molecular mechanisms governing the diverse phenotypes of S. aureus, ranging from a harmless colonizer to an organism capable of infecting various human organs. Emphasis will be placed on QS and its role in orchestrating S. aureus behavior across different contexts. SHORT CONCLUSION The pathophysiology of S. aureus infection is substantially influenced by phenotypic changes resulting from factors beyond Agr. Future studies are expected to give the comprehensive understanding of S. aureus overall profile in various settings.
Collapse
Affiliation(s)
- Yuriko Yamazaki
- Cutaneous Allergy and Host Defense, Immunology Frontier Research Center, Osaka, University, Osaka, 565-0871, Japan
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Tomoka Ito
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Masakazu Tamai
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Seitaro Nakagawa
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Yuumi Nakamura
- Cutaneous Allergy and Host Defense, Immunology Frontier Research Center, Osaka, University, Osaka, 565-0871, Japan.
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan.
| |
Collapse
|
29
|
Puengel T, Tacke F. Role of Kupffer cells and other immune cells. SINUSOIDAL CELLS IN LIVER DISEASES 2024:483-511. [DOI: 10.1016/b978-0-323-95262-0.00024-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
30
|
Kwak A, Thanabalasuriar A. Intravital Microscopy for Imaging and Live Cell Tracking of Alveolar Macrophages in Real Time. Methods Mol Biol 2024; 2813:189-204. [PMID: 38888779 DOI: 10.1007/978-1-0716-3890-3_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Classic in vitro coculture assays of pathogens with host cells have contributed significantly to our understanding of the intracellular lifestyle of several pathogens. Coculture assays with pathogens and eukaryotic cells can be analyzed through various techniques including plating for colony-forming units (CFU), confocal microscopy, and flow cytometry. However, findings from in vitro assays require validation in an in vivo model. Several physiological conditions can influence host-pathogen interactions, which cannot easily be mimicked in vitro. Intravital microscopy (IVM) is emerging as a powerful tool for studying host-pathogen interactions by enabling in vivo imaging of living organisms. As a result, IVM has significantly enhanced the understanding of infection mediated by diverse pathogens. The versatility of IVM has also allowed for the imaging of various organs as sites of local infection. This chapter specifically focuses on IVM conducted on the lung for elucidating pulmonary immune response, primarily involving alveolar macrophages, to pathogens. Additionally, in this chapter we outline the protocol for lung IVM that utilizes a thoracic suction window to stabilize the lung for acquiring stable images.
Collapse
Affiliation(s)
- Ashley Kwak
- School of Biomedical Sciences Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada
| | - Ajitha Thanabalasuriar
- School of Biomedical Sciences Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada.
| |
Collapse
|
31
|
Wang J, Dong D, Zhao W, Wang J. Intravital microscopy visualizes innate immune crosstalk and function in tissue microenvironment. Eur J Immunol 2024; 54:e2350458. [PMID: 37830252 DOI: 10.1002/eji.202350458] [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: 03/01/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/14/2023]
Abstract
Significant advances have been made in the field of intravital microscopy (IVM) on myeloid cells due to the growing number of validated fluorescent probes and reporter mice. IVM provides a visualization platform to directly observe cell behavior and deepen our understanding of cellular dynamics, heterogeneity, plasticity, and cell-cell communication in native tissue environments. This review outlines the current studies on the dynamic interaction and function of innate immune cells with a focus on those that are studied with IVM and covers the advances in data analysis with emerging artificial intelligence-based algorithms. Finally, the prospects of IVM on innate immune cells are discussed.
Collapse
Affiliation(s)
- Jin Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dong Dong
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenying Zhao
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Immune-related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
32
|
Goormaghtigh F, Van Bambeke F. Understanding Staphylococcus aureus internalisation and induction of antimicrobial tolerance. Expert Rev Anti Infect Ther 2024; 22:87-101. [PMID: 38180805 DOI: 10.1080/14787210.2024.2303018] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/04/2024] [Indexed: 01/07/2024]
Abstract
INTRODUCTION Staphylococcus aureus, a human commensal, is also one of the most common and serious pathogens for humans. In recent years, its capacity to survive and replicate in phagocytic and non-phagocytic cells has been largely demonstrated. In these intracellular niches, bacteria are shielded from the immune response and antibiotics, turning host cells into long-term infectious reservoirs. Moreover, neutrophils carry intracellular bacteria in the bloodstream, leading to systemic spreading of the disease. Despite the serious threat posed by intracellular S. aureus to human health, the molecular mechanisms behind its intracellular survival and subsequent antibiotic treatment failure remain elusive. AREA COVERED We give an overview of the killing mechanisms of phagocytes and of the impressive arsenal of virulence factors, toxins and stress responses deployed by S. aureus as a response. We then discuss the different barriers to antibiotic activity in this intracellular niche and finally describe innovative strategies to target intracellular persisting reservoirs. EXPERT OPINION Intracellular niches represent a challenge in terms of diagnostic and treatment. Further research using ad-hoc in-vivo models and single cell approaches are needed to better understand the molecular mechanisms underlying intracellular survival and tolerance to antibiotics in order to identify strategies to eliminate these persistent bacteria.
Collapse
Affiliation(s)
- Frédéric Goormaghtigh
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Françoise Van Bambeke
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
33
|
Wang J, An H, Ding M, Liu Y, Wang S, Jin Q, Wu Q, Dong H, Guo Q, Tian X, Liu J, Zhang J, Zhu T, Li J, Shao Z, Briles DE, Veening JW, Zheng H, Zhang L, Zhang JR. Liver macrophages and sinusoidal endothelial cells execute vaccine-elicited capture of invasive bacteria. Sci Transl Med 2023; 15:eade0054. [PMID: 38117903 DOI: 10.1126/scitranslmed.ade0054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 11/29/2023] [Indexed: 12/22/2023]
Abstract
Vaccination has substantially reduced the morbidity and mortality of bacterial diseases, but mechanisms of vaccine-elicited pathogen clearance remain largely undefined. We report that vaccine-elicited immunity against invasive bacteria mainly operates in the liver. In contrast to the current paradigm that migrating phagocytes execute vaccine-elicited immunity against blood-borne pathogens, we found that invasive bacteria are captured and killed in the liver of vaccinated host via various immune mechanisms that depend on the protective potency of the vaccine. Vaccines with relatively lower degrees of protection only activated liver-resident macrophage Kupffer cells (KCs) by inducing pathogen-binding immunoglobulin M (IgM) or low amounts of IgG. IgG-coated pathogens were directly captured by KCs via multiple IgG receptors FcγRs, whereas IgM-opsonized bacteria were indirectly bound to KCs via complement receptors of immunoglobulin superfamily (CRIg) and complement receptor 3 (CR3) after complement C3 activation at the bacterial surface. Conversely, the more potent vaccines engaged both KCs and liver sinusoidal endothelial cells by inducing higher titers of functional IgG antibodies. Endothelial cells (ECs) captured densely IgG-opsonized pathogens by the low-affinity IgG receptor FcγRIIB in a "zipper-like" manner and achieved bacterial killing predominantly in the extracellular milieu via an undefined mechanism. KC- and endothelial cell-based capture of antibody-opsonized bacteria also occurred in FcγR-humanized mice. These vaccine protection mechanisms in the liver not only provide a comprehensive explanation for vaccine-/antibody-boosted immunity against invasive bacteria but also may serve as in vivo functional readouts of vaccine efficacy.
Collapse
Affiliation(s)
- Juanjuan Wang
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Haoran An
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ming Ding
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yanhong Liu
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Shaomeng Wang
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Qian Jin
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Qi Wu
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Haodi Dong
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qile Guo
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xianbin Tian
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | | | | | - Tao Zhu
- Cansino Biologics, Tianjin 300301, China
| | | | - Zhujun Shao
- National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102299, China
| | - David E Briles
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Jan-Willem Veening
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne 1015, Switzerland
| | | | - Linqi Zhang
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Jing-Ren Zhang
- Center for Infectious Disease Research, Department of Basic Medical Science, School of Medicine, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
34
|
Kratofil RM. Working up an appetite to promote repair. Science 2023; 382:780. [PMID: 37972169 DOI: 10.1126/science.adl4292] [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: 11/19/2023]
Abstract
Immune-derived hunger hormones restore tissue after infection.
Collapse
Affiliation(s)
- Rachel M Kratofil
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
35
|
Slavetinsky J, Lehmann E, Slavetinsky C, Gritsch L, van Dalen R, Kretschmer D, Bleul L, Wolz C, Weidenmaier C, Peschel A. Wall Teichoic Acid Mediates Staphylococcus aureus Binding to Endothelial Cells via the Scavenger Receptor LOX-1. ACS Infect Dis 2023; 9:2133-2140. [PMID: 37910786 DOI: 10.1021/acsinfecdis.3c00252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
The success of Staphylococcus aureus as a major cause for endovascular infections depends on effective interactions with blood-vessel walls. We have previously shown that S. aureus uses its wall teichoic acid (WTA), a surface glycopolymer, to attach to endothelial cells. However, the endothelial WTA receptor remained unknown. We show here that the endothelial oxidized low-density lipoprotein receptor 1 (LOX-1) interacts with S. aureus WTA and permits effective binding of S. aureus to human endothelial cells. Purified LOX-1 bound to isolated S. aureus WTA. Ectopic LOX-1 expression led to increased binding of S. aureus wild type but not of a WTA-deficient mutant to a cell line, and LOX-1 blockage prevented S. aureus binding to endothelial cells. Moreover, WTA and LOX-1 expression levels correlated with the efficacy of the S. aureus-endothelial interaction. Thus, LOX-1 is an endothelial ligand for S. aureus, whose blockage may help to prevent or treat severe endovascular infections.
Collapse
Affiliation(s)
- Jessica Slavetinsky
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen72076, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen 72076, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen 72076 , Germany
| | - Esther Lehmann
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen72076, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen 72076, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen 72076 , Germany
| | - Christoph Slavetinsky
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen72076, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen 72076, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen 72076 , Germany
- Pediatric Surgery and Urology, University Children's Hospital Tübingen, Tübingen 72076, Germany
| | - Lisa Gritsch
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen72076, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen 72076, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen 72076 , Germany
| | - Rob van Dalen
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen72076, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen 72076, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen 72076 , Germany
| | - Dorothee Kretschmer
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen72076, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen 72076, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen 72076 , Germany
| | - Lisa Bleul
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen72076, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen 72076, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen 72076 , Germany
| | - Christiane Wolz
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen72076, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen 72076, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen 72076 , Germany
| | - Christopher Weidenmaier
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen72076, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen 72076, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen 72076 , Germany
| | - Andreas Peschel
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen72076, Germany
- Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen 72076, Germany
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen 72076 , Germany
| |
Collapse
|
36
|
Friot A, Djebali S, Valsesia S, Parroche P, Dubois M, Baude J, Vandenesch F, Marvel J, Leverrier Y. Antigen specific activation of cytotoxic CD8 + T cells by Staphylococcus aureus infected dendritic cells. Front Cell Infect Microbiol 2023; 13:1245299. [PMID: 37953797 PMCID: PMC10639145 DOI: 10.3389/fcimb.2023.1245299] [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: 06/23/2023] [Accepted: 09/22/2023] [Indexed: 11/14/2023] Open
Abstract
Staphylococcus aureus (S. aureus) is a pathogen associated with a wide variety of diseases, from minor to life-threatening infections. Antibiotic-resistant strains have emerged, leading to increasing concern about the control of S. aureus infections. The development of vaccines may be one way to overcome these resistant strains. However, S. aureus ability to internalize into cells - and thus to form a reservoir escaping humoral immunity - is a challenge for vaccine development. A role of T cells in the elimination of persistent S. aureus has been established in mice but it remains to be established if CD8+ T cells could display a cytotoxic activity against S. aureus infected cells. We examined in vitro the ability of CD8+ T cells to recognize and kill dendritic cells infected with S. aureus. We first evidenced that both primary mouse dendritic cells and DC2.4 cell line can be infected with S. aureus. We then generated a strain of S. aureus expressing a model CD8 epitope and transgenic F5 CD8+ T cells recognizing this model epitope were used as reporter T cells. In response to S. aureus-infected dendritic cells, F5 CD8+ T cells produced IFN-γ in an antigen-specific manner and displayed an increased ability to kill infected cells. Altogether, these results demonstrate that cells infected by S. aureus display bacteria-derived epitopes at their surface that are recognized by CD8+ T cells. This paves the way for the development of CD8+ T cell-based therapies against S. aureus.
Collapse
|
37
|
Dutta P, Bishayi B. Pyrrolidine dithiocarbamate in combination with L-N-monomethyl arginine alleviates Staphylococcus aureus infection via regulation of CXCL8/CXCR1 axis in peritoneal macrophages in vitro. Microb Pathog 2023; 183:106294. [PMID: 37567327 DOI: 10.1016/j.micpath.2023.106294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/07/2023] [Accepted: 08/07/2023] [Indexed: 08/13/2023]
Abstract
The CXCL8/CXCR1 axis in conjoint with the free radicals and anti-oxidants dictates the severity of inflammation caused by the bacteria, Staphylococcus aureus. S.aureus mediated inflammatory processes is regulated by NF-κB and its product, iNOS. The objective of this study was to examine the effects of inhibition of NF-κB and iNOS on CXCL8/CXCR1, alteration in M1/M2 polarization of macrophages and associated inflammatory responses during S.aureus infection in vitro. For this, the murine peritoneal macrophages were pretreated with NF-κB inhibitor, Pyrrolidine dithiocarbamate (PDTC) and iNOS inhibitor, L-N-monomethyl arginine (LNMMA), either alone or in combination, followed by time-dependent S.aureus infection. The chemotactic migrations of macrophages were determined by the agarose spot assay. The iNOS, NF-κB and CXCR1 protein expressions were evaluated. The ROS level (superoxide, H2O2, NO) and antioxidant activities (SOD, CAT, GSH, arginase) were measured. The intra-macrophage phagoctyic activity had been analyzed by confocal microscopy. S.aureus activated macrophages showed increased iNOS expression that symbolizes M1 characterization of macrophages. The results suggest that the combination treatment of LNMMA + PDTC was effective in diminution of CXCL8 production and CXCR1 expression through downregulation of NF-κB and iNOS signaling pathway. Consequently, there was decrement in macrophage migration, reduced ROS generation, elevated antioxidant enzyme activity as well as bacterial phagocytosis at 90 min post bacterial infection. The increased arginase activity further proves the switch from pro-inflammatory M1 to anti-inflammatory M2 polarization of macrophages. Concludingly, the combination of PDTC + LNMMA could resolve S.aureus mediated inflammation through mitigation of CXCL8/CXCR1 pathway switching from M1 to M2 polarization.
Collapse
Affiliation(s)
- Puja Dutta
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta, 700009, West Bengal, India
| | - Biswadev Bishayi
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta, 700009, West Bengal, India.
| |
Collapse
|
38
|
Peiseler M, Araujo David B, Zindel J, Surewaard BGJ, Lee WY, Heymann F, Nusse Y, Castanheira FVS, Shim R, Guillot A, Bruneau A, Atif J, Perciani C, Ohland C, Ganguli Mukherjee P, Niehrs A, Thuenauer R, Altfeld M, Amrein M, Liu Z, Gordon PMK, McCoy K, Deniset J, MacParland S, Ginhoux F, Tacke F, Kubes P. Kupffer cell-like syncytia replenish resident macrophage function in the fibrotic liver. Science 2023; 381:eabq5202. [PMID: 37676943 DOI: 10.1126/science.abq5202] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/13/2023] [Indexed: 09/09/2023]
Abstract
Kupffer cells (KCs) are localized in liver sinusoids but extend pseudopods to parenchymal cells to maintain their identity and serve as the body's central bacterial filter. Liver cirrhosis drastically alters vascular architecture, but how KCs adapt is unclear. We used a mouse model of liver fibrosis and human tissue to examine immune adaptation. Fibrosis forced KCs to lose contact with parenchymal cells, down-regulating "KC identity," which rendered them incapable of clearing bacteria. Commensals stimulated the recruitment of monocytes through CD44 to a spatially distinct vascular compartment. There, recruited monocytes formed large aggregates of multinucleated cells (syncytia) that expressed phenotypical KC markers and displayed enhanced bacterial capture ability. Syncytia formed via CD36 and were observed in human cirrhosis as a possible antimicrobial defense that evolved with fibrosis.
Collapse
Affiliation(s)
- Moritz Peiseler
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow Klinikum and Campus Charité Mitte, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Bruna Araujo David
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Joel Zindel
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Visceral Surgery and Medicine, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Bas G J Surewaard
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Woo-Yong Lee
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Felix Heymann
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Ysbrand Nusse
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Fernanda V S Castanheira
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Raymond Shim
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Adrien Guillot
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Alix Bruneau
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Jawairia Atif
- Ajmera Transplant Centre, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Catia Perciani
- Ajmera Transplant Centre, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Christina Ohland
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Annika Niehrs
- Leibniz Institute of Virology (LIV), Hamburg, Germany
| | | | | | - Mathias Amrein
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Paul M K Gordon
- Centre for Health Genomics and Informatics, University of Calgary, Calgary, Alberta, Canada
| | - Kathy McCoy
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Justin Deniset
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Cardiac Sciences, University of Calgary, Calgary, Alberta, Canada
- Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sonya MacParland
- Ajmera Transplant Centre, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Gustave Roussy Cancer Campus, INSERM U1015, Villejuif, France
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Paul Kubes
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
39
|
Loredan DG, Devlin JC, Lacey KA, Howard N, Chen Z, Zwack EE, Lin JD, Ruggles KV, Khanna KM, Torres VJ, Loke P. Single-Cell Analysis of CX3CR1+ Cells Reveals a Pathogenic Role for BIRC5+ Myeloid Proliferating Cells Driven by Staphylococcus aureus Leukotoxins. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:836-843. [PMID: 37466391 PMCID: PMC10450158 DOI: 10.4049/jimmunol.2300166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/30/2023] [Indexed: 07/20/2023]
Abstract
Our previous studies identified a population of stem cell-like proliferating myeloid cells within inflamed tissues that could serve as a reservoir for tissue macrophages to adopt different activation states depending on the microenvironment. By lineage-tracing cells derived from CX3CR1+ precursors in mice during infection and profiling by single-cell RNA sequencing, in this study, we identify a cluster of BIRC5+ myeloid cells that expanded in the liver during chronic infection with either the parasite Schistosoma mansoni or the bacterial pathogen Staphylococcus aureus. In the absence of tissue-damaging toxins, S. aureus infection does not elicit these BIRC5+ cells. Moreover, deletion of BIRC5 from CX3CR1-expressing cells results in improved survival during S. aureus infection. Hence the combination of single-cell RNA sequencing and genetic fate-mapping CX3CR1+ cells revealed a toxin-dependent pathogenic role for BIRC5 in myeloid cells during S. aureus infection.
Collapse
Affiliation(s)
- Denis G. Loredan
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
- Vilcek Institute of Graduate Biomedical Sciences, New York University Grossman School of Medicine, New York, NY
| | - Joseph C. Devlin
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| | - Keenan A. Lacey
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| | - Nina Howard
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Ze Chen
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| | - Erin E. Zwack
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| | - Jian-Da Lin
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei City, Taiwan
- Center for Computational and Systems Biology, National Taiwan University, Taipei City, Taiwan
| | - Kelly V. Ruggles
- Institute of Systems Genetics, New York University Grossman School of Medicine, New York, NY
- Division of Precision Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Kamal M. Khanna
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
- Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY
| | - P’ng Loke
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
40
|
Yang S, Zeng J, Hao W, Sun R, Tuo Y, Tan L, Zhang H, Liu R, Bai H. IL-21/IL-21R Promotes the Pro-Inflammatory Effects of Macrophages during C. muridarum Respiratory Infection. Int J Mol Sci 2023; 24:12557. [PMID: 37628738 PMCID: PMC10454239 DOI: 10.3390/ijms241612557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Interleukin-21 and its receptors (IL-21/IL-21R) aggravate chlamydial lung infection, while macrophages (Mφ) are one of the main cells infected by chlamydia and the main source of inflammatory cytokines. Therefore, it is particularly important to study whether IL-21/IL-21R aggravates chlamydia respiratory infection by regulating Mφ. Combined with bioinformatics analysis, we established an IL-21R-deficient (IL-21R-/-) mouse model of Chlamydia muridarum (C. muridarum) respiratory tract infection in vivo, studied C. muridarum-stimulated RAW264.7 by the addition of rmIL-21 in vitro, and conducted adoptive transfer experiments to clarify the association between IL-21/IL-21R and Mφ. IL-21R-/- mice showed lower infiltration of pulmonary total Mφ, alveolar macrophages, and interstitial macrophages compared with WT mice following infection. Transcriptomic analysis suggested that M1-related genes are downregulated in IL-21R-/- mice and that IL-21R deficiency affects the Mφ-mediated inflammatory response during C. muridarum infection. In vivo experiments verified that in IL-21R-/- mice, pulmonary M1-type CD80+, CD86+, MHC II+, TNFα+, and iNOS+ Mφ decreased, while there were no differences in M2-type CD206+, TGF-β+, IL-10+ and ARG1+ Mφ. In vitro, administration of rmIL-21 to C. muridarum-stimulated RAW264.7 cells promoted the levels of iNOS-NO and the expression of IL-12p40 and TNFα, but had no effect on TGFβ or IL-10. Further, adoptive transfer of M1-like bone marrow-derived macrophages derived from IL-21R-/- mice, unlike those from WT mice, effectively protected the recipients against C. muridarum infection and induced relieved pulmonary pathology. These findings help in understanding the mechanism by which IL-21/IL-21R exacerbates chlamydia respiratory infection by promoting the proinflammatory effect of Mφ.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hong Bai
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; (S.Y.); (J.Z.); (W.H.); (R.S.); (Y.T.); (L.T.); (H.Z.); (R.L.)
| |
Collapse
|
41
|
Sriyanti C, Siregar TN, Mudatsir M, Gani A, Hasan DI, Sutriana A. Antibacterial and anti-inflammatory activities of Nothopanax scutellarium, Moringa oleifera and Piper betle extracts on staphylococcal mastitis animal model. NARRA J 2023; 3:e176. [PMID: 38454978 PMCID: PMC10919737 DOI: 10.52225/narra.v3i2.176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/07/2023] [Indexed: 03/09/2024]
Abstract
Inappropriate and prolonged administration of antibiotics in mastitis could cause resistance and herbal treatment might could be one alternative treatment. Nothopanax scutellarium, Moringa oleifera, and Piper betle are medicinal plants that contain various active compounds, including antibacterial and anti-inflammatory agents, but their potential in treating mastitis is minimum. The aim of this study was to assess the effectiveness of those plants against mastitis in rabbit model induced by Staphylococcus aureus. A total of 25 lactating rabbits (Oryctolagus cuniculus) weighing 3.0±0.4 kg were grouped into five groups: healthy control; mastitis control, and three treatment groups (Nothopanax scutellarium, Moringa oleifera, and Piper betle). Except the negative control, all animals were inoculated with 0.15 mL of S. aureus containing 1.5x107 colony forming unit (CFU)/mL on eight days after giving birth. The extract was administered orally after four hours Staphylococcus aureus inoculation at a dose of 50 mg/kg body weight, twice a day for five consecutive days. The number of bacteria in the milk and the level of serum interleukin 6 (IL-6) were measured and histopathological examination of mammary gland tissues were analyzed. The log number of total plate count of Staphylococcus aureus indicated that all extract groups had significant lower of bacterial logs compared to mastitis control (all comparisons had p<0.05) with the lowest was found in Piper betle group, followed by Nothopanax scutellarium and Moringa oleifera groups. The enzyme-linked immunosorbent assay (ELISA) results showed that all ethanolic extract groups had significantly lower levels of IL-6 compared to the mastitis control (all comparisons had p<0.05). The histopathology assessment suggested that extract groups had lower infiltration of inflammatory cells such as lymphocytes and macrophages in alveoli compared to the mastitis control group. In conclusion, all three extracts contained antibacterial and anti-inflammatory activities and Piper betle had the most effective in reducing bacterial growth and IL-6 level compared to others.
Collapse
Affiliation(s)
- Cut Sriyanti
- Graduate School of Mathematics and Applied Sciences, Universitas Syiah Kuala, Banda Aceh, Indonesia
- Department of Midwifery, Health Polytechnic of Aceh Ministry of Health, Aceh Besar, Indonesia
| | - Tongku N. Siregar
- Laboratory of Reproduction, Faculty of Veterinary Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Mudatsir Mudatsir
- Department of Microbiology, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Azhari Gani
- Department of Internal Medicine, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Denny I. Hasan
- Laboratory of Pathology, Faculty of Veterinary Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Amalia Sutriana
- Laboratory of Pharmacology, Faculty of Veterinary Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| |
Collapse
|
42
|
Pinto RM, Yazdani S, Seabra CL, De Jonge M, Izci M, Cruz R, Casal S, Soenen SJ, Reis S, Nunes C, Van Dijck P. Non disseminative nano-strategy against in vivo Staphylococcus aureus biofilms. NPJ Biofilms Microbiomes 2023; 9:39. [PMID: 37328504 DOI: 10.1038/s41522-023-00405-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/05/2023] [Indexed: 06/18/2023] Open
Abstract
Staphylococcus aureus is considered a high priority pathogen by the World Health Organization due to its high prevalence and the potential to form biofilms. Currently, the available treatments for S. aureus biofilm-associated infections do not target the extracellular polymeric substances (EPS) matrix. This matrix is a physical barrier to bactericidal agents, contributing to the increase of antimicrobial tolerance. The present work proposes the development of lipid nanoparticles encapsulating caspofungin (CAS) as a matrix-disruptive nanosystem. The nanoparticles were functionalized with D-amino acids to target the matrix. In a multi-target nano-strategy against S. aureus biofilms, CAS-loaded nanoparticles were combined with a moxifloxacin-loaded nanosystem, as an adjuvant to promote the EPS matrix disruption. In vitro and in vivo studies showed biofilm reduction after combining the two nanosystems. Besides, the combinatory therapy showed no signs of bacterial dissemination into vital organs of mice, while dissemination was observed for the treatment with the free compounds. Additionally, the in vivo biodistribution of the two nanosystems revealed their potential to reach and accumulate in the biofilm region, after intraperitoneal administration. Thus, this nano-strategy based on the encapsulation of matrix-disruptive and antibacterial agents is a promising approach to fight S. aureus biofilms.
Collapse
Affiliation(s)
- Rita M Pinto
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, 4050-313, Porto, Portugal
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, 3001, Leuven, Belgium
| | - Saleh Yazdani
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, 3001, Leuven, Belgium
| | - Catarina Leal Seabra
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, 4050-313, Porto, Portugal
| | - Martine De Jonge
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, 3001, Leuven, Belgium
| | - Mukaddes Izci
- NanoHealth and Optical Imaging Group, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Rebeca Cruz
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, 4050-313, Porto, Portugal
| | - Susana Casal
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, 4050-313, Porto, Portugal
| | - Stefaan J Soenen
- NanoHealth and Optical Imaging Group, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Salette Reis
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, 4050-313, Porto, Portugal
| | - Cláudia Nunes
- LAQV, REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, 4050-313, Porto, Portugal.
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, 3001, Leuven, Belgium.
| |
Collapse
|
43
|
Chen Z, Xie Z, Han M, Jin Q, Li Z, Zhai Y, Zhang M, Hu G, Zhang H. Global Transcriptomic Study of Circular-RNA Expression Profile in Osteoclasts Infected by Intracellular Staphylococcus aureus. Infect Immun 2023; 91:e0035722. [PMID: 37212691 PMCID: PMC10269070 DOI: 10.1128/iai.00357-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 05/01/2023] [Indexed: 05/23/2023] Open
Abstract
Osteomyelitis is difficult to cure, and the rapidly rising morbidity is a thorny problem accompanied by a large number of joint replacement applications. Staphylococcus aureus is the main pathogen of osteomyelitis. Circular RNAs (circRNAs), as emerging noncoding RNAs, play important roles in multiple physiopathological processes which could provide novel insights into osteomyelitis. However, little is known about the roles of circRNAs in the pathogenesis of osteomyelitis. Osteoclasts, considered bone sentinels, are the resident macrophages in bone and may play the immune defense roles in osteomyelitis. It has been reported that S. aureus can survive in osteoclasts, but the function of osteoclast circRNAs in response to intracellular S. aureus infection remains unclear. In this study, we investigated the profile of circRNAs in osteoclasts infected by intracellular S. aureus through high-throughput RNA sequencing. In total, 24 upregulated and 62 downregulated differentially expressed circRNAs were identified and subsequently analyzed to demonstrate their potential functions. On this basis, three circRNAs (chr4:130718154-130728164+, chr8:77409548-77413627-, and chr1:190871592-190899571-) were confirmed as potential novel biomarkers for the diagnosis of osteomyelitis through the murine model of osteomyelitis. Most importantly, we verified that the circRNA chr4:130718154-130728164+ named circPum1 could regulate the host autophagy to affect the intracellular infection of S. aureus through miR-767. In addition, circPum1 could serve as a promising serum biomarker in osteomyelitis patients caused by S. aureus infection. Taken together, this study provided the first global transcriptomic profile analysis of circRNAs in osteoclasts infected by intracellular S. aureus and first proposed a novel perspective for the pathogenesis and immunotherapy of S. aureus-induced osteomyelitis from the term of circRNAs.
Collapse
Affiliation(s)
- Zhihao Chen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Zonggang Xie
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Mingxiao Han
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Qiyuan Jin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Ziyuan Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Yaxuan Zhai
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Minxing Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Gangfeng Hu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Haifang Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| |
Collapse
|
44
|
Andrade S, Ramalho MJ, Santos SB, Melo LDR, Santos RS, Guimarães N, Azevedo NF, Loureiro JA, Pereira MC. Fighting Methicillin-Resistant Staphylococcus aureus with Targeted Nanoparticles. Int J Mol Sci 2023; 24:ijms24109030. [PMID: 37240376 DOI: 10.3390/ijms24109030] [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: 04/11/2023] [Revised: 05/03/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Antimicrobial resistance (AMR) is considered one of the greatest threats to global health. Methicillin-resistant Staphylococcus aureus (MRSA) remains at the core of this threat, accounting for about 90% of S. aureus infections widespread in the community and hospital settings. In recent years, the use of nanoparticles (NPs) has emerged as a promising strategy to treat MRSA infections. NPs can act directly as antibacterial agents via antibiotic-independent activity and/or serve as drug delivery systems (DDSs), releasing loaded antibiotics. Nonetheless, directing NPs to the infection site is fundamental for effective MRSA treatment so that highly concentrated therapeutic agents are delivered to the infection site while directly reducing the toxicity to healthy human cells. This leads to decreased AMR emergence and less disturbance of the individual's healthy microbiota. Hence, this review compiles and discusses the scientific evidence related to targeted NPs developed for MRSA treatment.
Collapse
Affiliation(s)
- Stéphanie Andrade
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Maria J Ramalho
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Sílvio B Santos
- CEB-Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
| | - Luís D R Melo
- CEB-Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal
- LABBELS-Associate Laboratory, University of Minho, 4710-057 Braga, Portugal
| | - Rita S Santos
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Nuno Guimarães
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Nuno F Azevedo
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Joana A Loureiro
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Maria C Pereira
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| |
Collapse
|
45
|
Qi X, Shen N, Al Othman A, Mezentsev A, Permyakova A, Yu Z, Lepoitevin M, Serre C, Durymanov M. Metal-Organic Framework-Based Nanomedicines for the Treatment of Intracellular Bacterial Infections. Pharmaceutics 2023; 15:1521. [PMID: 37242762 PMCID: PMC10220673 DOI: 10.3390/pharmaceutics15051521] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Metal-organic frameworks (MOFs) are a highly versatile class of ordered porous materials, which hold great promise for different biomedical applications, including antibacterial therapy. In light of the antibacterial effects, these nanomaterials can be attractive for several reasons. First, MOFs exhibit a high loading capacity for numerous antibacterial drugs, including antibiotics, photosensitizers, and/or photothermal molecules. The inherent micro- or meso-porosity of MOF structures enables their use as nanocarriers for simultaneous encapsulation of multiple drugs resulting in a combined therapeutic effect. In addition to being encapsulated into an MOF's pores, antibacterial agents can sometimes be directly incorporated into an MOF skeleton as organic linkers. Next, MOFs contain coordinated metal ions in their structure. Incorporation of Fe2/3+, Cu2+, Zn2+, Co2+, and Ag+ can significantly increase the innate cytotoxicity of these materials for bacteria and cause a synergistic effect. Finally, abundance of functional groups enables modifying the external surface of MOF particles with stealth coating and ligand moieties for improved drug delivery. To date, there are a number of MOF-based nanomedicines available for the treatment of bacterial infections. This review is focused on biomedical consideration of MOF nano-formulations designed for the therapy of intracellular infections such as Staphylococcus aureus, Mycobacterium tuberculosis, and Chlamydia trachomatis. Increasing knowledge about the ability of MOF nanoparticles to accumulate in a pathogen intracellular niche in the host cells provides an excellent opportunity to use MOF-based nanomedicines for the eradication of persistent infections. Here, we discuss advantages and current limitations of MOFs, their clinical significance, and their prospects for the treatment of the mentioned infections.
Collapse
Affiliation(s)
- Xiaoli Qi
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Ningfei Shen
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Aya Al Othman
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | | | | | - Zhihao Yu
- Institute of Porous Materials from Paris (IMAP), Ecole Normale Supérieure, ESPCI Paris, CNRS, PSL University, 75006 Paris, France
| | - Mathilde Lepoitevin
- Institute of Porous Materials from Paris (IMAP), Ecole Normale Supérieure, ESPCI Paris, CNRS, PSL University, 75006 Paris, France
| | - Christian Serre
- Institute of Porous Materials from Paris (IMAP), Ecole Normale Supérieure, ESPCI Paris, CNRS, PSL University, 75006 Paris, France
| | - Mikhail Durymanov
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
- Faculty of Chemistry, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
46
|
Zucoloto AZ, Schlechte J, Ignacio A, Thomson CA, Pyke S, Yu IL, Geuking MB, McCoy KD, Yipp BG, Gillrie MR, McDonald B. Vascular traffic control of neutrophil recruitment to the liver by microbiota-endothelium crosstalk. Cell Rep 2023; 42:112507. [PMID: 37195866 DOI: 10.1016/j.celrep.2023.112507] [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: 11/21/2022] [Revised: 03/20/2023] [Accepted: 04/28/2023] [Indexed: 05/19/2023] Open
Abstract
During bloodstream infections, neutrophils home to the liver as part of an intravascular immune response to eradicate blood-borne pathogens, but the mechanisms regulating this crucial response are unknown. Using in vivo imaging of neutrophil trafficking in germ-free and gnotobiotic mice, we demonstrate that the intestinal microbiota guides neutrophil homing to the liver in response to infection mediated by the microbial metabolite D-lactate. Commensal-derived D-lactate augments neutrophil adhesion in the liver independent of granulopoiesis in bone marrow or neutrophil maturation and activation in blood. Instead, gut-to-liver D-lactate signaling primes liver endothelial cells to upregulate adhesion molecule expression in response to infection and promote neutrophil adherence. Targeted correction of microbiota D-lactate production in a model of antibiotic-induced dysbiosis restores neutrophil homing to the liver and reduces bacteremia in a model of Staphylococcus aureus infection. These findings reveal long-distance traffic control of neutrophil recruitment to the liver by microbiota-endothelium crosstalk.
Collapse
Affiliation(s)
- Amanda Z Zucoloto
- Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jared Schlechte
- Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Aline Ignacio
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Carolyn A Thomson
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Shannon Pyke
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Ian-Ling Yu
- Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Markus B Geuking
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kathy D McCoy
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Bryan G Yipp
- Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mark R Gillrie
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Braedon McDonald
- Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
47
|
Buckley PT, Chan R, Fernandez J, Luo J, Lacey KA, DuMont AL, O'Malley A, Brezski RJ, Zheng S, Malia T, Whitaker B, Zwolak A, Payne A, Clark D, Sigg M, Lacy ER, Kornilova A, Kwok D, McCarthy S, Wu B, Morrow B, Nemeth-Seay J, Petley T, Wu S, Strohl WR, Lynch AS, Torres VJ. Multivalent human antibody-centyrin fusion protein to prevent and treat Staphylococcus aureus infections. Cell Host Microbe 2023; 31:751-765.e11. [PMID: 37098341 DOI: 10.1016/j.chom.2023.04.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 02/12/2023] [Accepted: 04/03/2023] [Indexed: 04/27/2023]
Abstract
Treating and preventing infections by antimicrobial-resistant bacterial pathogens is a worldwide problem. Pathogens such as Staphylococcus aureus produce an array of virulence determinants, making it difficult to identify single targets for the development of vaccines or monoclonal therapies. We described a human-derived anti-S. aureus monoclonal antibody (mAb)-centyrin fusion protein ("mAbtyrin") that simultaneously targets multiple bacterial adhesins, resists proteolysis by bacterial protease GluV8, avoids Fc engagement by S. aureus IgG-binding proteins SpA and Sbi, and neutralizes pore-forming leukocidins via fusion with anti-toxin centyrins, while maintaining Fc- and complement-mediated functions. Compared with the parental mAb, mAbtyrin protected human phagocytes and boosted phagocyte-mediated killing. The mAbtyrin also reduced pathology, reduced bacterial burden, and protected from different types of infections in preclinical animal models. Finally, mAbtyrin synergized with vancomycin, enhancing pathogen clearance in an animal model of bacteremia. Altogether, these data establish the potential of multivalent mAbs for treating and preventing S. aureus diseases.
Collapse
Affiliation(s)
- Peter T Buckley
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA.
| | - Rita Chan
- Department of Microbiology, New York University Grossman School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Jeffrey Fernandez
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Jinquan Luo
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Keenan A Lacey
- Department of Microbiology, New York University Grossman School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Ashley L DuMont
- Department of Microbiology, New York University Grossman School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Aidan O'Malley
- Department of Microbiology, New York University Grossman School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Randall J Brezski
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Songmao Zheng
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Thomas Malia
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Brian Whitaker
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Adam Zwolak
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Angela Payne
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Desmond Clark
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Martin Sigg
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Eilyn R Lacy
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Anna Kornilova
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Debra Kwok
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Steve McCarthy
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Bingyuan Wu
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Brian Morrow
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | | | - Ted Petley
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - Sam Wu
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | - William R Strohl
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, USA
| | | | - Victor J Torres
- Department of Microbiology, New York University Grossman School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA; Antimicrobial-Resistant Pathogens Program, New York University Langone Health, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA.
| |
Collapse
|
48
|
Parsons JB, Westgeest AC, Conlon BP, Fowler VG. Persistent Methicillin-Resistant Staphylococcus aureus Bacteremia: Host, Pathogen, and Treatment. Antibiotics (Basel) 2023; 12:455. [PMID: 36978320 PMCID: PMC10044482 DOI: 10.3390/antibiotics12030455] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/18/2023] [Accepted: 02/20/2023] [Indexed: 03/02/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a devastating pathogen responsible for a variety of life-threatening infections. A distinctive characteristic of this pathogen is its ability to persist in the bloodstream for several days despite seemingly appropriate antibiotics. Persistent MRSA bacteremia is common and is associated with poor clinical outcomes. The etiology of persistent MRSA bacteremia is a result of the complex interplay between the host, the pathogen, and the antibiotic used to treat the infection. In this review, we explore the factors related to each component of the host-pathogen interaction and discuss the clinical relevance of each element. Next, we discuss the treatment options and diagnostic approaches for the management of persistent MRSA bacteremia.
Collapse
Affiliation(s)
- Joshua B. Parsons
- Department of Medicine, Division of Infectious Disease, Duke University Medical Center, Durham, NC 27710, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Annette C. Westgeest
- Department of Medicine, Division of Infectious Disease, Duke University Medical Center, Durham, NC 27710, USA
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Brian P. Conlon
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Vance G. Fowler
- Department of Medicine, Division of Infectious Disease, Duke University Medical Center, Durham, NC 27710, USA
- Duke Clinical Research Institute, Durham, NC 27710, USA
| |
Collapse
|
49
|
A Sequalae of Lineage Divergence in Staphylococcus aureus from Community-Acquired Patterns in Youth to Hospital-Associated Profiles in Seniors Implied Age-Specific Host-Selection from a Common Ancestor. Diagnostics (Basel) 2023; 13:diagnostics13050819. [PMID: 36899963 PMCID: PMC10001379 DOI: 10.3390/diagnostics13050819] [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: 01/30/2023] [Revised: 02/10/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
The rapidly changing epidemiology of Staphylococcus aureus and evolution of strains with enhanced virulence is a significant issue in global healthcare. Hospital-associated methicillin-resistant S. aureus (HA-MRSA) lineages are being completely replaced by community-associated S. aureus (CA-MRSA) in many regions. Surveillance programs tracing the reservoirs and sources of infections are needed. Using molecular diagnostics, antibiograms, and patient demographics, we have examined the distributions of S. aureus in Ha'il hospitals. Out of 274 S. aureus isolates recovered from clinical specimens, 181 (66%, n = 181) were MRSA, some with HA-MRSA patterns across 26 antimicrobials with almost full resistances to all beta-lactams, while the majority were highly susceptible to all non-beta-lactams, indicating the CA-MRSA type. The rest of isolates (34%, n = 93) were methicillin-susceptible, penicillin-resistant MSSA lineages (90%). The MRSA in men was over 56% among total MRSA (n = 181) isolates and 37% of overall isolates (n = 102 of 274) compared to MSSA in total isolates (17.5%, n = 48), respectively. However, these were 28.4% (n = 78) and 12.4% (n = 34) for MRSA and MSSA infections in women, respectively. MRSA rates per age groups of 0-20, 21-50, and >50 years of age were 15% (n = 42), 17% (n = 48), and 32% (n = 89), respectively. However, MSSA in the same age groups were 13% (n = 35), 9% (n = 25), and 8% (n = 22). Interestingly, MRSA increased proportional to age, while MSSA concomitantly decreased, implying dominance of the latter ancestors early in life and then gradual replacement by MRSA. The dominance and seriousness of MRSA despite enormous efforts in place is potentially for the increased use of beta-lactams known to enhance virulence. The Intriguing prevalence of the CA-MRSA patterns in young otherwise healthy individuals replaced by MRSA later in seniors and the dominance of penicillin-resistant MSSA phenotypes imply three types of host- and age-specific evolutionary lineages. Thus, the decreasing MSSA trend by age with concomitant increase and sub-clonal differentiation into HA-MRSA in seniors and CA-MRSA in young and otherwise healthy patients strongly support the notion of subclinal emergences from a resident penicillin-resistant MSSA ancestor. Future vertical studies should focus on the surveillance of invasive CA-MRSA rates and phenotypes.
Collapse
|
50
|
MRSA Isolates from Patients with Persistent Bacteremia Generate Nonstable Small Colony Variants In Vitro within Macrophages and Endothelial Cells during Prolonged Vancomycin Exposure. Infect Immun 2023; 91:e0042322. [PMID: 36602380 PMCID: PMC9872686 DOI: 10.1128/iai.00423-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Staphylococcus aureus (especially methicillin-resistant S. aureus [MRSA]) is frequently associated with persistent bacteremia (PB) during vancomycin therapy despite consistent susceptibility in vitro. Strategic comparisons of PB strains versus those from vancomycin-resolving bacteremia (RB) would yield important mechanistic insights into PB outcomes. Clinical PB versus RB isolates were assessed in vitro for intracellular replication and small colony variant (SCV) formation within macrophages and endothelial cells (ECs) in the presence or absence of exogenous vancomycin. In both macrophages and ECs, PB and RB isolates replicated within lysosome-associated membrane protein-1 (LAMP-1)-positive compartments. PB isolates formed nonstable small colony variants (nsSCVs) in vancomycin-exposed host cells at a significantly higher frequency than matched RB isolates (in granulocyte-macrophage colony-stimulating factor [GM-CSF], human macrophages PB versus RB, P < 0.0001 at 48 h; in ECs, PB versus RB, P < 0.0001 at 24 h). This phenotype could represent one potential basis for the unique ability of PB isolates to adaptively resist vancomycin therapy and cause PB in humans. Elucidating the molecular mechanism(s) by which PB strains form nsSCVs could facilitate the discovery of novel treatment strategies to mitigate PB due to MRSA.
Collapse
|