1
|
Xiao K, Cao Y, Han Z, Zhang Y, Luu LDW, Chen L, Yan P, Chen W, Wang J, Liang Y, Shi X, Wang X, Wang F, Hu Y, Wen Z, Chen Y, Yang Y, Yu H, Xie L, Wang Y. A pan-immune panorama of bacterial pneumonia revealed by a large-scale single-cell transcriptome atlas. Signal Transduct Target Ther 2025; 10:5. [PMID: 39757231 DOI: 10.1038/s41392-024-02093-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 01/07/2025] Open
Abstract
Bacterial pneumonia is a significant public health burden, contributing to substantial morbidity, mortality, and healthcare costs. Current therapeutic strategies beyond antibiotics and adjuvant therapies are limited, highlighting the need for a deeper understanding of the disease pathogenesis. Here, we employed single-cell RNA sequencing of 444,146 bronchoalveolar lavage fluid cells (BALFs) from a large cohort of 74 individuals, including 58 patients with mild (n = 22) and severe (n = 36) diseases as well as 16 healthy donors. Enzyme-linked immunosorbent and histological assays were applied for validation within this cohort. The heterogeneity of immune responses in bacterial pneumonia was observed, with distinct immune cell profiles related to disease severity. Severe bacterial pneumonia was marked by an inflammatory cytokine storm resulting from systemic upregulation of S100A8/A9 and CXCL8, primarily due to specific macrophage and neutrophil subsets. In contrast, mild bacterial pneumonia exhibits an effective humoral immune response characterized by the expansion of T follicular helper and T helper 2 cells, facilitating B cell activation and antibody production. Although both disease groups display T cell exhaustion, mild cases maintained robust cytotoxic CD8+T cell function, potentially reflecting a compensatory mechanism. Dysregulated neutrophil and macrophage responses contributed significantly to the pathogenesis of severe disease. Immature neutrophils promote excessive inflammation and suppress T cell activation, while a specific macrophage subset (Macro_03_M1) displaying features akin to myeloid-derived suppressor cells (M-MDSCs) suppress T cells and promote inflammation. Together, these findings highlight potential therapeutic targets for modulating immune responses and improving clinical outcomes in bacterial pneumonia.
Collapse
Affiliation(s)
- Kun Xiao
- College of Pulmonary & Critical Care Medicine, The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100091, P.R. China.
| | - Yan Cao
- College of Pulmonary & Critical Care Medicine, The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100091, P.R. China
| | - Zhihai Han
- College of Pulmonary & Critical Care Medicine, The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100091, P.R. China
| | - Yuxiang Zhang
- Department of Critical Care Medicine, The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100037, P.R. China
| | - Laurence Don Wai Luu
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, 2007, Australia
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Liang Chen
- Respiratory and Critical Care Medicine department, Beijing Jingmei Group, General Hospial, Beijing, 102308, P.R. China
| | - Peng Yan
- Department of Pulmonary and Critical Care Medicine, China Aerospace Science & Industry Corporation 731 hospital, Beijing, 100074, P.R. China
| | - Wei Chen
- College of Pulmonary & Critical Care Medicine, The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100091, P.R. China
- Department of Respiratory Medicine, The Sixth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, 100037, P.R. China
| | - Jiaxing Wang
- Department of Critical Care Medicine, The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100037, P.R. China
| | - Ying Liang
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, P.R. China
| | - Xin Shi
- College of Pulmonary & Critical Care Medicine, The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100091, P.R. China
- Medical School of Chinese PLA, Beijing, 100191, P.R. China
| | - Xiuli Wang
- College of Pulmonary & Critical Care Medicine, The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100091, P.R. China
- Medical School of Chinese PLA, Beijing, 100191, P.R. China
| | - Fan Wang
- College of Pulmonary & Critical Care Medicine, The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100091, P.R. China
| | - Ye Hu
- College of Pulmonary & Critical Care Medicine, The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100091, P.R. China
| | - Zhengjun Wen
- Respiratory and Critical Care Medicine department, Beijing Jingmei Group, General Hospial, Beijing, 102308, P.R. China
| | - Yong Chen
- Department of Pulmonary and Critical Care Medicine, Anzhen hospital afflicted to Capital medical university, Beijing, 100029, P.R. China
| | - Yuwei Yang
- College of Pulmonary & Critical Care Medicine, The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100091, P.R. China
| | - Haotian Yu
- The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100091, P.R. China.
| | - Lixin Xie
- College of Pulmonary & Critical Care Medicine, The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100091, P.R. China.
| | - Yi Wang
- Experimental Research Center, Capital Institute of Pediatrics, Beijing, 100020, P.R. China.
| |
Collapse
|
2
|
Dou MH, Huang JY, Li PY, Chen WL, Wang XR, Yang TZ, Fan XY, Zhang XY, Lu Y, Bai J, Du SY. How can traditional Chinese medicine enhance the efficacy of antibiotics in the treatment of MRSA-infected pneumonia: An experimental study on the combination of Reyanning mixture (RYN) and linezolid. JOURNAL OF ETHNOPHARMACOLOGY 2024; 340:119221. [PMID: 39653103 DOI: 10.1016/j.jep.2024.119221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Reyanning Mixture (RYN) is a Chinese patent medicine widely used in the treatment of respiratory inflammatory diseases in China and has potential in the treatment of bacteria-infected pneumonia. AIM OF THE STUDY The present study aimed to demonstrate the therapeutic potential of RYN in combination with linezolid for the treatment of MRSA-infected pneumonia and to explore the mechanisms of action and active components. MATERIALS AND METHODS The pharmacodynamics of RYN alone and in combination with linezolid was investigated in a rat model of MRSA-induced pneumonia. Transcriptomics, ELISA, Western blot and qRT-PCR were used to explore and verify the pharmacological mechanism of the anti-inflammatory effect of RYN. UPLC-MS and molecular docking were used to explore the anti-inflammatory components of RYN for the treatment of MRSA-infected pneumonia. RESULTS In vivo, RYN reduced lung injury and inflammation in rats with pneumonia. In particular, the combination of RYN and linezolid enhanced the therapeutic effect compared to that of either treatment alone. Further research suggests that the synergistic therapeutic effect of the combination may be related to the inhibition of the inflammatory response by RYN and the enhancement of linezolid inhibition and clearance of MRSA in lung tissues by RYN. RYN plays an anti-inflammatory role in MRSA-infected pneumonia by inhibiting the TLR2/NF-κB/NLRP3 pathway, with 7 active components that may play a dominant role. CONCLUSIONS These results indicate that RYN may serve as an adjuvant drug to antibiotics for the treatment of MRSA-associated pneumonia. Exploration of its mechanisms and active components is conducive to the clinical application and quality improvement of RYN. More importantly, this study showed that the synergistic therapeutic effect of the combination of traditional Chinese medicine and antibiotics may be a valuable therapeutic strategy.
Collapse
Affiliation(s)
- Min-Hang Dou
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jia-Yi Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Peng-Yue Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wan-Ling Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xin-Ran Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Tian-Zi Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiao-Yu Fan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xin-Yu Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yang Lu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Jie Bai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Shou-Ying Du
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
3
|
Zelasko S, Swaney MH, Sandstrom S, Lee KE, Dixon J, Riley C, Watson L, Godfrey JJ, Ledrowski N, Rey F, Safdar N, Seroogy CM, Gern JE, Kalan L, Currie C. Early-life upper airway microbiota are associated with decreased lower respiratory tract infections. J Allergy Clin Immunol 2024:S0091-6749(24)01189-8. [PMID: 39547283 DOI: 10.1016/j.jaci.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Microbial interactions mediating colonization resistance play key roles within the human microbiome, shaping susceptibility to infection from birth. The role of the nasal and oral microbiome in the context of early life respiratory infections and subsequent allergic disease risk remains understudied. OBJECTIVES Our aim was to gain insight into microbiome-mediated defenses and respiratory pathogen colonization dynamics within the upper respiratory tract during infancy. METHODS We performed shotgun metagenomic sequencing of nasal (n = 229) and oral (n = 210) microbiomes from our Wisconsin Infant Study Cohort at age 24 months and examined the influence of participant demographics and exposure history on microbiome composition. Detection of viral and bacterial respiratory pathogens by RT-PCR and culture-based studies with antibiotic susceptibility testing, respectively, to assess pathogen carriage was performed. Functional bioassays were used to evaluate pathogen inhibition by respiratory tract commensals. RESULTS Participants with early-life lower respiratory tract infection were more likely to be formula fed, attend day care, and experience wheezing. Composition of the nasal, but not oral, microbiome associated with prior lower respiratory tract infection, namely lower alpha diversity, depletion of Prevotella, and enrichment of Moraxella catarrhalis including drug-resistant strains. Prevotella originating from healthy microbiomes had higher biosynthetic gene cluster abundance and exhibited contact-independent inhibition of M catarrhalis. CONCLUSIONS These results suggest interbacterial competition affects nasal pathogen colonization. This work advances understanding of protective host-microbe interactions occurring in airway microbiomes that alter infection susceptibility in early life.
Collapse
Affiliation(s)
- Susan Zelasko
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wis; Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wis.
| | - Mary Hannah Swaney
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wis; Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wis
| | - Shelby Sandstrom
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wis
| | - Kristine E Lee
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wis
| | - Jonah Dixon
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Colleen Riley
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Lauren Watson
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Jared J Godfrey
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Naomi Ledrowski
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Federico Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wis
| | - Nasia Safdar
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis; William S. Middleton Memorial Veterans Affairs Hospital, Madison, Wis
| | - Christine M Seroogy
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - James E Gern
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis; Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Lindsay Kalan
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wis; Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis; M. G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Cameron Currie
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wis; M. G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
4
|
Dong H, Zhao Y, Li S, Wang Q, Li M, Zhao K, Zhang Z, Shi J, Zhao X, Liu J. An inhalable nanoparticle enabling virulence factor elimination and antibiotics delivery for pneumococcal pneumonia therapy. J Control Release 2024; 375:698-711. [PMID: 39313100 DOI: 10.1016/j.jconrel.2024.09.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 09/06/2024] [Accepted: 09/20/2024] [Indexed: 09/25/2024]
Abstract
Streptococcus pneumoniae (S. pneumoniae) is a major cause of community-acquired pneumonia. Current standard clinical therapies mainly focus on combating S. pneumoniae through antibiotics. However, the limited delivery of antibiotics and the undetoxified hydrogen peroxide (H2O2) virulence factor secreted by S. pneumoniae impede the therapeutic outcomes. Here we report an inhalable catalase (CAT)-tannic acid (TA) nanoassembly for local antibiotic (levofloxacin) delivery and simultaneously neutralizing the secreted H2O2 virulence factors to treat pneumococcal pneumonia. After aerosol inhalation, the inhalable formulation (denoted as CT@LVX) effectively accumulates in lung tissues through TA-mediated mucoadhesion. CAT can reduce alveolar epithelial cells apoptosis by catalyzing the decomposition of accumulated H2O2 in the infected lung tissues. In synergy with antibiotic LVX-mediated S. pneumoniae elimination, CT@LVX significantly decreases lung injury companied with reduced inflammatory, resulting in 100 % survival of mice with pneumonia. In a clinically isolated S. pneumoniae strain-induced pneumonia mouse model, CT@LVX also shows superior outcomes compared to the traditional antibiotic treatment, highlighting its potential clinical application prospects.
Collapse
Affiliation(s)
- Huiyue Dong
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yuxin Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Shihong Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, PR China
| | - Qiongwei Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Mengli Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Kaikai Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Xiu Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Junjie Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
5
|
Rago F, Melo EM, Miller LM, Duray AM, Batista Felix F, Vago JP, de Faria Gonçalves AP, Angelo ALPM, Cassali GD, de Gaetano M, Brennan E, Owen B, Guiry P, Godson C, Alcorn JF, Teixeira MM. Treatment with lipoxin A 4 improves influenza A infection outcome, induces macrophage reprogramming, anti-inflammatory and pro-resolutive responses. Inflamm Res 2024; 73:1903-1918. [PMID: 39214890 DOI: 10.1007/s00011-024-01939-9] [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: 05/28/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
INTRODUCTION Influenza A is a virus from the Orthomixoviridae family responsible for high lethality rates and morbidity, despite clinically proven vaccination strategies and some anti-viral therapies. The eicosanoid Lipoxin A4 (LXA4) promotes the resolution of inflammation by decreasing cell recruitment and pro-inflammatory cytokines release, but also for inducing activation of apoptosis, efferocytosis, and macrophage reprogramming. OBJECTIVE Here, we evaluated whether a synthetic lipoxin mimetic, designated AT-01-KG, would improve the course of influenza A infection in a murine model. METHOD Mice were infected with influenza A/H1N1 and treated with AT-01-KG (1.7 μg/kg/day, i.p.) at day 3 post-infection. RESULTS AT-01-KG attenuated mortality, reducing leukocyte infiltration and lung damage at day 5 and day 7 post-infection. AT-01-KG is a Formyl Peptide Receptor 2 (designated FPR2/3 in mice) agonist, and the protective responses were not observed in fpr2/3 -/- animals. In mice treated with LXA4 (50 μg/kg/day, i.p., days 3-6 post-infection), at day 7, macrophage reprogramming was observed, as seen by a decrease in classically activated macrophages and an increase in alternatively activated macrophages in the lungs. Furthermore, the number of apoptotic cells and cells undergoing efferocytosis was increased in the lavage of treated mice. Treatment also modulated the adaptive immune response, increasing the number of T helper 2 cells (Th2) and regulatory T (Tregs) cells in the lungs of the treated mice. CONCLUSION Therefore, treatment with a lipoxin A4 analog was beneficial in a model of influenza A infection in mice. The drug decreased inflammation and promoted resolution and beneficial immune responses, suggesting it may be useful in patients with severe influenza.
Collapse
Affiliation(s)
- Flavia Rago
- Department of Biochemistry and Immunology Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, CEP 31.270-901, Belo Horizonte, MG, 6627, Brazil.
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, 9127 Rangos Research Building, 4401 Penn Ave, Pittsburgh, PA, 15224, USA.
| | - Eliza Mathias Melo
- Department of Biochemistry and Immunology Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, CEP 31.270-901, Belo Horizonte, MG, 6627, Brazil
| | - Leigh M Miller
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, 9127 Rangos Research Building, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Alexis M Duray
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, 9127 Rangos Research Building, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Franciel Batista Felix
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Juliana Priscila Vago
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana Paula de Faria Gonçalves
- Immunology of Viral Diseases, René Rachou Research Center, Oswaldo Cruz Foundation (FIOCRUZ-Minas), Belo Horizonte, MG, Brazil
| | | | - Geovanni D Cassali
- Comparative Pathology Laboratory, Department of Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Monica de Gaetano
- School of Medicine/School of Biomolecular and Biomedical Science, UCD Diabetes Complications Research Centre, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Eoin Brennan
- School of Medicine/School of Biomolecular and Biomedical Science, UCD Diabetes Complications Research Centre, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Benjamin Owen
- Centre for Synthesis and Chemical Biology, School of Chemistry, University College Dublin, Dublin, Ireland
| | - Patrick Guiry
- Centre for Synthesis and Chemical Biology, School of Chemistry, University College Dublin, Dublin, Ireland
| | - Catherine Godson
- School of Medicine/School of Biomolecular and Biomedical Science, UCD Diabetes Complications Research Centre, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - John F Alcorn
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, 9127 Rangos Research Building, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Avenida Antônio Carlos, CEP 31.270-901, Belo Horizonte, MG, 6627, Brazil.
| |
Collapse
|
6
|
Dou L, Wang X, Bai Y, Li Q, Luo L, Yu W, Wang Z, Wen K, Shen J. Mussel-like polydopamine-assisted aggregation-induced emission nanodot for enhanced broad-spectrum antimicrobial activity: In vitro and in vivo validation. Int J Biol Macromol 2024; 282:136762. [PMID: 39486741 DOI: 10.1016/j.ijbiomac.2024.136762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/22/2024] [Accepted: 10/19/2024] [Indexed: 11/04/2024]
Abstract
Emerging luminogens with aggregation-induced emission properties, namely AIEgens, demonstrated excellent anti-bacteria activity potential. However, their application still limited by the low antibacterial activity caused by the poor binding with bacteria. Polydopamine (PDA), an important biological macromolecule, possesses superior adhesion ability toward various material surface, including bacteria. In this study, the novel mussel-like PDA-assisted AIE Nanodot was proposed, achieving with robust bacterial binding ability and enhanced broad-spectrum antibacterial activity. Binding ability inherited from the PDA enables the constructed PDA-assisted AIE Nanodot to adhere efficiently to the bacterial membrane surface. Meanwhile, the AIE properties endowed them with monitoring capability, allowing for tracking their interaction with bacteria through facile fluorescence imaging in real time. More importantly, excellent killing of both Gram-positive and Gram-negative bacteria were successfully achieved in vitro antibacterial tests with excellent biocompatibility. Furthermore, in the treatment of Methicillin-resistant S. aureus (MRSA)-infected mouse-wound model, the mice exhibited accelerated wound healing with low bacterial load. This novel integrated strategy introduced a simple but effectively design to enhance the binding and antibacterial ability of AIEgens and would diversify the existing pool of antibacterial agents.
Collapse
Affiliation(s)
- Leina Dou
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China; College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shanxi, China
| | - Xiaonan Wang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Yuchen Bai
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Qing Li
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Liang Luo
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Wenbo Yu
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Zhanhui Wang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Kai Wen
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China.
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China.
| |
Collapse
|
7
|
Sanches Santos Rizzo Zuttion M, Parimon T, Bora SA, Yao C, Lagree K, Gao CA, Wunderink RG, Kitsios GD, Morris A, Zhang Y, McVerry BJ, Modes ME, Marchevsky AM, Stripp BR, Soto CM, Wang Y, Merene K, Cho S, Victor BL, Vujkovic-Cvijin I, Gupta S, Cassel SL, Sutterwala FS, Devkota S, Underhill DM, Chen P. Antibiotic use during influenza infection augments lung eosinophils that impair immunity against secondary bacterial pneumonia. J Clin Invest 2024; 134:e180986. [PMID: 39255040 PMCID: PMC11527449 DOI: 10.1172/jci180986] [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: 03/12/2024] [Accepted: 09/06/2024] [Indexed: 09/12/2024] Open
Abstract
A leading cause of mortality after influenza infection is the development of a secondary bacterial pneumonia. In the absence of a bacterial superinfection, prescribing antibacterial therapies is not indicated but has become a common clinical practice for those presenting with a respiratory viral illness. In a murine model, we found that antibiotic use during influenza infection impaired the lung innate immunologic defenses toward a secondary challenge with methicillin-resistant Staphylococcus aureus (MRSA). Antibiotics augment lung eosinophils, which have inhibitory effects on macrophage function through the release of major basic protein. Moreover, we demonstrated that antibiotic treatment during influenza infection caused a fungal dysbiosis that drove lung eosinophilia and impaired MRSA clearance. Finally, we evaluated 3 cohorts of hospitalized patients and found that eosinophils positively correlated with antibiotic use, systemic inflammation, and worsened outcomes. Altogether, our work demonstrates a detrimental effect of antibiotic treatment during influenza infection that has harmful immunologic consequences via recruitment of eosinophils to the lungs, thereby increasing the risk of developing a secondary bacterial infection.
Collapse
Affiliation(s)
| | | | | | - Changfu Yao
- Department of Medicine
- Women’s Guild Lung Institute
| | - Katherine Lagree
- Department of Biomedical Sciences
- Widjaja Foundation Inflammatory Bowel Disease Institute, and
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Catherine A. Gao
- Division of Pulmonary and Critical Care, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Richard G. Wunderink
- Division of Pulmonary and Critical Care, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Georgios D. Kitsios
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine and
- Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alison Morris
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine and
- Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yingze Zhang
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine and
| | - Bryan J. McVerry
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine and
- Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | - Barry R. Stripp
- Department of Medicine
- Women’s Guild Lung Institute
- Department of Biomedical Sciences
| | | | - Ying Wang
- Department of Medicine
- Women’s Guild Lung Institute
| | | | - Silvia Cho
- Department of Medicine
- Women’s Guild Lung Institute
| | | | - Ivan Vujkovic-Cvijin
- Department of Medicine
- Department of Biomedical Sciences
- Widjaja Foundation Inflammatory Bowel Disease Institute, and
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Suman Gupta
- Department of Medicine
- Women’s Guild Lung Institute
| | | | | | - Suzanne Devkota
- Department of Medicine
- Department of Biomedical Sciences
- Widjaja Foundation Inflammatory Bowel Disease Institute, and
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Human Microbiome Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - David M. Underhill
- Department of Biomedical Sciences
- Widjaja Foundation Inflammatory Bowel Disease Institute, and
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Peter Chen
- Department of Medicine
- Women’s Guild Lung Institute
- Department of Biomedical Sciences
| |
Collapse
|
8
|
Yang S, Tong T, Wang H, Li Z, Wang M, Ni K. Causal relationship between air pollution and infections: a two-sample Mendelian randomization study. Front Public Health 2024; 12:1409640. [PMID: 39148655 PMCID: PMC11324489 DOI: 10.3389/fpubh.2024.1409640] [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: 03/30/2024] [Accepted: 07/16/2024] [Indexed: 08/17/2024] Open
Abstract
Background Traditional observational studies exploring the association between air pollution and infections have been limited by small sample sizes and potential confounding factors. To address these limitations, we applied Mendelian randomization (MR) to investigate the potential causal relationships between particulate matter (PM2.5, PM2.5-10, and PM10), nitrogen dioxide, and nitrogen oxide and the risks of infections. Methods Single nucleotide polymorphisms (SNPs) related to air pollution were selected from the genome-wide association study (GWAS) of the UK Biobank. Publicly available summary data for infections were obtained from the FinnGen Biobank and the COVID-19 Host Genetics Initiative. The inverse variance weighted (IVW) meta-analysis was used as the primary method for obtaining the Mendelian randomization (MR) estimates. Complementary analyses were performed using the weighted median method, MR-Egger method, and MR Pleiotropy Residual Sum and Outlier (MR-PRESSO) test. Results The fixed-effect IVW estimate showed that PM2.5, PM2.5-10 and Nitrogen oxides were suggestively associated with COVID-19 [for PM2.5: IVW (fe): OR 3.573(1.218,5.288), PIVW(fe) = 0.021; for PM2.5-10: IVW (fe): OR 2.940(1.385,6.239), PIVW(fe) = 0.005; for Nitrogen oxides, IVW (fe): OR 1.898(1.318,2.472), PIVW(fe) = 0.010]. PM2.5, PM2.5-10, PM10, and Nitrogen oxides were suggestively associated with bacterial pneumonia [for PM2.5: IVW(fe): OR 1.720 (1.007, 2.937), PIVW(fe) = 0.047; for PM2.5-10: IVW(fe): OR 1.752 (1.111, 2.767), P IVW(fe) = 0.016; for PM10: IVW(fe): OR 2.097 (1.045, 4.208), PIVW(fe) = 0.037; for Nitrogen oxides, IVW(fe): OR 3.907 (1.209, 5.987), PIVW(fe) = 0.023]. Furthermore, Nitrogen dioxide was suggestively associated with the risk of acute upper respiratory infections, while all air pollution were not associated with intestinal infections. Conclusions Our results support a role of related air pollution in the Corona Virus Disease 2019, bacterial pneumonia and acute upper respiratory infections. More work is need for policy formulation to reduce the air pollution and the emission of toxic and of harmful gas.
Collapse
Affiliation(s)
- Shengyi Yang
- Department of Infection Control, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tong Tong
- Department of Infection Control, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hong Wang
- Department of Infection Control, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhenwei Li
- Department of Infection Control, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Mengmeng Wang
- Department of Infection Control, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Kaiwen Ni
- Department of Infection Control, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
9
|
Zhang J, Qi H, Wu JJ, Mao X, Zhang H, Amin N, Xu F, Dong C, Wang C, Wang P, Zheng L. Disposable Peptidoglycan-Specific Biosensor for Noninvasive Real-Time Detection of Broad-Spectrum Gram-Positive Bacteria in Exhaled Breath Condensates. Anal Chem 2024; 96:9817-9825. [PMID: 38730304 DOI: 10.1021/acs.analchem.4c00059] [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: 05/12/2024]
Abstract
Rapidly identifying and quantifying Gram-positive bacteria are crucial to diagnosing and treating bacterial lower respiratory tract infections (LRTIs). This work presents a field-deployable biosensor for detecting Gram-positive bacteria from exhaled breath condensates (EBCs) based on peptidoglycan recognition using an aptamer. Dielectrophoretic force is employed to enrich the bacteria in 10 s without additional equipment or steps. Concurrently, the measurement of the sensor's interfacial capacitance is coupled to quantify the bacteria during the enrichment process. By incorporation of a semiconductor condenser, the whole detection process, including EBC collection, takes about 3 min. This biosensor has a detection limit of 10 CFU/mL, a linear range of up to 105 CFU/mL and a selectivity of 1479:1. It is cost-effective and disposable due to its low cost. The sensor provides a nonstaining, culture-free and PCR-independent solution for noninvasive and real-time diagnosis of Gram-positive bacterial LRTIs.
Collapse
Affiliation(s)
- Jian Zhang
- College of Electrical and Electronic Engineering, Wenzhou University, Wenzhou 325035, China
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China
| | - Haochen Qi
- College of Electrical and Electronic Engineering, Wenzhou University, Wenzhou 325035, China
- Department of Electrical Engineering and Computer Science, the University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Jie Jayne Wu
- Department of Electrical Engineering and Computer Science, the University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Xuanjiao Mao
- Clinical Laboratory, The People's Hospital of Pingyang, Wenzhou 325400, China
| | - Hailin Zhang
- Department of Children's Respiratory Medicine, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Niloufar Amin
- Department of Electrical Engineering and Computer Science, the University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Feng Xu
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
| | - Changkun Dong
- College of Electrical and Electronic Engineering, Wenzhou University, Wenzhou 325035, China
| | - Chunchang Wang
- Laboratory of Dielectric Functional Materials, School of Materials Science & Engineering, Anhui University, Hefei 230601, China
| | - Pengjun Wang
- College of Electrical and Electronic Engineering, Wenzhou University, Wenzhou 325035, China
| | - Lei Zheng
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| |
Collapse
|
10
|
Rago F, Melo EM, Miller LM, Duray AM, Felix FB, Vago JP, Gonçalves APF, Angelo ALPM, Cassali GD, Gaetano M, Brennan E, Owen B, Guiry P, Godson C, Alcorn JF, Teixeira MM. Treatment with lipoxin A 4 improves influenza A infection outcome through macrophage reprogramming, anti-inflammatory and pro-resolutive responses. RESEARCH SQUARE 2024:rs.3.rs-4491036. [PMID: 38947034 PMCID: PMC11213203 DOI: 10.21203/rs.3.rs-4491036/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Objective and design Here, we evaluated whether a synthetic lipoxin mimetic, designated AT-01-KG, would improve the course of influenza A infection in a murine model. Treatment Mice were infected with influenza A/H1N1 and treated with AT-01-KG (1.7 mg/kg/day, i.p.) at day 3 post-infection. Methods Mortality rate was assessed up to day 21 and inflammatory parameters were assessed at days 5 and 7. Results AT-01-KG attenuated mortality, reducing leukocyte infiltration and lung damage at day 5 and day 7 post-infection. AT-01-KG is a Formyl Peptide Receptor 2 (designated FPR2/3 in mice) agonist, and the protective responses were not observed in FPR2/3 -/- animals. In mice treated with LXA4 (50mg/kg/day, i.p., days 3-6 post-infection), at day 7, macrophage reprogramming was observed, as seen by a decrease in classically activated macrophages and an increase in alternatively activated macrophages in the lungs. Furthermore, the number of apoptotic cells and cells undergoing efferocytosis was increased in the lavage of treated mice. Treatment also modulated the adaptive immune response, increasing the number of anti-inflammatory T cells (Th2) and regulatory T (Tregs) cells in the lungs of the treated mice. Conclusions Therefore, treatment with a lipoxin A4 analog was beneficial in a model of influenza A infection in mice. The drug decreased inflammation and promoted resolution and beneficial immune responses, suggesting it may be useful in patients with severe influenza.
Collapse
|
11
|
Kouroupis PC, O'Rourke N, Kelly S, McKittrick M, Noppe E, Reyes LF, Rodriguez A, Martin-Loeches I. Hospital-acquired bacterial pneumonia in critically ill patients: from research to clinical practice. Expert Rev Anti Infect Ther 2024; 22:423-433. [PMID: 38743435 DOI: 10.1080/14787210.2024.2354828] [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: 02/26/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024]
Abstract
INTRODUCTION Hospital-acquired pneumonia (HAP) represents a significant cause of mortality among critically ill patients admitted to Intensive Care Units (ICUs). Timely and precise diagnosis is imperative to enhance therapeutic efficacy and patient outcomes. However, the diagnostic process is challenged by test limitations and a wide-ranging list of differential diagnoses, particularly in patients exhibiting escalating oxygen requirements, leukocytosis, and increased secretions. AREAS COVERED This narrative review aims to update diagnostic modalities, facilitating the prompt identification of nosocomial pneumonia while guiding, developing, and assessing therapeutic interventions. A comprehensive literature review was conducted utilizing the MEDLINE/PubMed database from 2013 to April 2024. EXPERT OPINION An integrated approach that integrates clinical, microbiological, and imaging tools is paramount. Progress in diagnostic techniques, including novel molecular methods, the expanding utilization and accuracy of bedside ultrasound, and the emergence of Artificial Intelligence, coupled with an improved comprehension of lung microbiota and host-pathogen interactions, continues to enhance our capability to accurately and swiftly identify HAP and its causative agents. This advancement enables the refinement of treatment strategies and facilitates the implementation of precision medicine approaches.
Collapse
Affiliation(s)
- Pompeo Costantino Kouroupis
- Department of Intensive Care Medicine, Multidisciplinary Intensive Care Research Organization (MICRO), St James' Hospital, Dublin, Ireland
| | - Niall O'Rourke
- Department of Intensive Care Medicine, Multidisciplinary Intensive Care Research Organization (MICRO), St James' Hospital, Dublin, Ireland
| | - Sinead Kelly
- Department of Intensive Care Medicine, Multidisciplinary Intensive Care Research Organization (MICRO), St James' Hospital, Dublin, Ireland
| | - Myles McKittrick
- Department of Intensive Care Medicine, Multidisciplinary Intensive Care Research Organization (MICRO), St James' Hospital, Dublin, Ireland
| | - Elne Noppe
- Department of Intensive Care Medicine, Multidisciplinary Intensive Care Research Organization (MICRO), St James' Hospital, Dublin, Ireland
| | - Luis F Reyes
- Department of Intensive Care Medicine, Unisabana Center for Translational Science, Chia, Colombia
- Department of Intensive Care Medicine, Clinica Universidad de La Sabana, Chia, Colombia
- Department of Intensive Care Medicine, Pandemic Sciences Institute, University of Oxford, Oxford, UK
| | - Alejandro Rodriguez
- Critical Care Department, Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain
- Department of Intensive Care Medicine, URV/IISPV/CIBERES, Tarragona, Spain
| | - Ignacio Martin-Loeches
- Department of Intensive Care Medicine, Multidisciplinary Intensive Care Research Organization (MICRO), St James' Hospital, Dublin, Ireland
- Hospital Clinic, Universitat de Barcelona, IDIBAPS, CIBERES, Barcelona, Spain
| |
Collapse
|
12
|
Yimiti M, Fei X, Yang H, Yang X, Li S, Tuoheniyazi H, Liu D, Ma J, Xie J, Zheng J, Song Z, Li Q, Xu D, Zhao Y, Gu Z. HDAC6 inhibitor promotes reactive oxygen species-meditated clearance of Staphylococcus aureus in macrophage. Clin Exp Pharmacol Physiol 2024; 51:e13866. [PMID: 38719209 DOI: 10.1111/1440-1681.13866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/02/2024] [Accepted: 04/06/2024] [Indexed: 05/30/2024]
Abstract
Staphylococcus aureus (S. aureus) pneumonia has become an increasingly important public health problem. Recent evidence suggests that epigenetic modifications are critical in the host immune defence against pathogen infection. In this study, we found that S. aureus infection induces the expression of histone deacetylase 6 (HDAC6) in a dose-dependent manner. Furthermore, by using a S. aureus pneumonia mouse model, we showed that the HDAC6 inhibitor, tubastatin A, demonstrates a protective effect in S. aureus pneumonia, decreasing the mortality and destruction of lung architecture, reducing the bacterial burden in the lungs and inhibiting inflammatory responses. Mechanistic studies in primary bone marrow-derived macrophages demonstrated that the HDAC6 inhibitors, tubastatin A and tubacin, reduced the intracellular bacterial load by promoting bacterial clearance rather than regulating phagocytosis. Finally, N-acetyl-L- cysteine, a widely used reactive oxygen species (ROS) scavenger, antagonized ROS production and significantly inhibited tubastatin A-induced S. aureus clearance. These findings demonstrate that HDAC6 inhibitors promote the bactericidal activity of macrophages by inducing ROS, an important host factor for S. aureus clearance and production. Our study identified HDAC6 as a suitable epigenetic modification target for preventing S. aureus infection, and tubastatin A as a useful compound in treating S. aureus pneumonia.
Collapse
Affiliation(s)
- Maimaitiaili Yimiti
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuefeng Fei
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Yang
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaobao Yang
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuhui Li
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huxidanmu Tuoheniyazi
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Danping Liu
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junrui Ma
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jialing Xie
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juanjuan Zheng
- Blood Transfusion Department, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong, China
| | - Zhen Song
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingtian Li
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dakang Xu
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanan Zhao
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Health Sciences and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhidong Gu
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Laboratory Medicine, Ruijin-Hainan Hospital, Shanghai Jiao Tong University School of Medicine (Hainan Boao Research Hospital), Qionghai, Hainan, China
| |
Collapse
|
13
|
Majumder S, Li P, Das S, Nafiz TN, Kumar S, Bai G, Dellario H, Sui H, Guan Z, Curtiss R, Furuya Y, Sun W. A bacterial vesicle-based pneumococcal vaccine against influenza-mediated secondary Streptococcus pneumoniae pulmonary infection. Mucosal Immunol 2024; 17:169-181. [PMID: 38215909 PMCID: PMC11033695 DOI: 10.1016/j.mucimm.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/29/2023] [Accepted: 01/08/2024] [Indexed: 01/14/2024]
Abstract
Streptococcus pneumoniae (Spn) is a common pathogen causing a secondary bacterial infection following influenza, which leads to severe morbidity and mortality during seasonal and pandemic influenza. Therefore, there is an urgent need to develop bacterial vaccines that prevent severe post-influenza bacterial pneumonia. Here, an improved Yersinia pseudotuberculosis strain (designated as YptbS46) possessing an Asd+ plasmid pSMV92 could synthesize high amounts of the Spn pneumococcal surface protein A (PspA) antigen and monophosphoryl lipid A as an adjuvant. The recombinant strain produced outer membrane vesicles (OMVs) enclosing a high amount of PspA protein (designated as OMV-PspA). A prime-boost intramuscular immunization with OMV-PspA induced both memory adaptive and innate immune responses in vaccinated mice, reduced the viral and bacterial burden, and provided complete protection against influenza-mediated secondary Spn infection. Also, the OMV-PspA immunization afforded significant cross-protection against the secondary Spn A66.1 infection and long-term protection against the secondary Spn D39 challenge. Our study implies that an OMV vaccine delivering Spn antigens can be a new promising pneumococcal vaccine candidate.
Collapse
Affiliation(s)
- Saugata Majumder
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Peng Li
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Shreya Das
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Tanvir Noor Nafiz
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Sudeep Kumar
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Guangchun Bai
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Hazel Dellario
- Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Haixin Sui
- Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Ziqiang Guan
- Department of Biochemistry, Duke University Medical Center, Durham, North Carolina, USA
| | - Roy Curtiss
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Yoichi Furuya
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA.
| | - Wei Sun
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA.
| |
Collapse
|
14
|
Xu Y, Bao L, Cao S, Pang B, Zhang J, Zhang Y, Chen M, Wang Y, Sun Q, Zhao R, Guo S, Sun J, Cui X. Pharmacological effects and mechanism of Maxing Shigan decoction in the treatment of Pseudomonas aeruginosa pneumonia. JOURNAL OF ETHNOPHARMACOLOGY 2024; 320:117424. [PMID: 37984543 DOI: 10.1016/j.jep.2023.117424] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/05/2023] [Accepted: 11/11/2023] [Indexed: 11/22/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Maxing Shigan Decoction (MXSG) is a traditional Chinese Medicine effectively used in respiratory infections and bacterial pneumonia. However, the mechanism of MXSG treating acute Pseudomonas aeruginosa (P. aeruginosa) pneumonia is still unclear. AIM OF THE STUDY This study aimed to investigate the therapeutic effects of MXSG on acute P. aeruginosa pneumonia and explore its potential mechanisms. MATERIALS AND METHODS HPLC-MS analysis was performed to analyze the chemical composition. Antibacterial effects in vitro were evaluated by minimum inhibitory concentration (MIC). Forty-five male BALB/c mice were divided into control group, model group, levofloxacin group, MXSG-L (7.7 g/kg/d), and MXSG-H group (15.4 g/kg/d). Mice were intranasal instillation with P. aeruginosa to induce acute P. aeruginosa pneumonia model. Levofloxacin and MXSG were administered by oral gavage once a day. After 3 days of treatment, the lung index measurement, micro-CT, arterial blood gas analysis, bacteria load determination, and HE staining were performed. Network pharmacological analysis and transcriptome sequencing were employed to predict the potential mechanisms of MXSG on bacterial pneumonia. The expressions of relating genes were detected by immunofluorescence, Western blot, and RT-PCR. RESULTS In vitro, MIC of P. aeruginosa is greater than 500 mg/mL. In the treatment of acute P. aeruginosa pneumonia model, MXSG significantly improved body weight loss, lung index, and pulmonary lesions. MXSG treatment also reduced the bacterial load and ameliorated oxygen saturation significantly. Transcriptomes, immunofluorescence, Western blot, and RT-PCR analysis showed MXSG treating acute P. aeruginosa pneumonia through the IL-17 signaling pathway and HIF-1α/IL-6/STAT3 signaling pathway. CONCLUSIONS We demonstrated the efficacy and mechanism of MXSG in the treatment of acute P. aeruginosa pneumonia, which provides a scientific basis for its clinical application.
Collapse
Affiliation(s)
- Yingli Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Lei Bao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Shan Cao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Bo Pang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Jingsheng Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Yu Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Mengping Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Yaxin Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Qiyue Sun
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Ronghua Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Shanshan Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Jing Sun
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Xiaolan Cui
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
15
|
Martins DM, Cardoso EM, Capellari L, Botelho LAB, Ferreira FA. Detection of Staphylococcus aureus from nares of elderly living in a Brazilian nursing home. Diagn Microbiol Infect Dis 2024; 108:116089. [PMID: 37931385 DOI: 10.1016/j.diagmicrobio.2023.116089] [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: 07/05/2023] [Revised: 08/23/2023] [Accepted: 09/14/2023] [Indexed: 11/08/2023]
Abstract
Asymptomatically nasal colonization by Staphylococcus aureus is a well-established risk factor for S. aureus infections. The aimed of the study was to identify the prevalence and factors associated with nasal carriage of S. aureus and Methicillin-resistant S. aureus (MRSA) from individuals residing in one Brazilian nursing home (NH). Three time-separate nasal swab collections were obtained from the elderly enrolled. The S. aureus isolates identified were submitted to Antimicrobial Susceptibility test (AST). The study showed a high prevalence of S. aureus (n = 9; 60%) and MRSA (n = 4; 26.7%) among elderly. Resistance to erythromycin was the most detected. S. aureus or MRSA colonization could not be associated to the data collected on demographics, personal habits, and medical history of the participants. Despite the small number of individuals enrolled, our study can contribute to improve the control of S. aureus and MRSA dissemination within the community, especially among the most vulnerable like the elderly.
Collapse
Affiliation(s)
- Damaris Miriã Martins
- Laboratório de Genética Molecular Bacteriana (GeMBac), Departamento de Microbiologia, Imunologia e Parasitologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Emanuela Mendes Cardoso
- Laboratório de Genética Molecular Bacteriana (GeMBac), Departamento de Microbiologia, Imunologia e Parasitologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Lilian Capellari
- Laboratório de Genética Molecular Bacteriana (GeMBac), Departamento de Microbiologia, Imunologia e Parasitologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Larissa Alvarenga Batista Botelho
- Departamento de Microbiologia Medica, Instituto de Microbiologia Paulo de Goes, Centro de Ciencias da Saude, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Fabienne Antunes Ferreira
- Laboratório de Genética Molecular Bacteriana (GeMBac), Departamento de Microbiologia, Imunologia e Parasitologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil.
| |
Collapse
|
16
|
Chen S, Jiang J, Li T, Huang L. PANoptosis: Mechanism and Role in Pulmonary Diseases. Int J Mol Sci 2023; 24:15343. [PMID: 37895022 PMCID: PMC10607352 DOI: 10.3390/ijms242015343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/09/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
PANoptosis is a newly defined programmed cell death (PCD) triggered by a series of stimuli, and it engages three well-learned PCD forms (pyroptosis, apoptosis, necroptosis) concomitantly. Normally, cell death is recognized as a strategy to eliminate unnecessary cells, inhibit the proliferation of invaded pathogens and maintain homeostasis; however, vigorous cell death can cause excessive inflammation and tissue damage. Acute lung injury (ALI) and chronic obstructive pulmonary syndrome (COPD) exacerbation is related to several pathogens (e.g., influenza A virus, SARS-CoV-2) known to cause PANoptosis. An understanding of the mechanism and specific regulators may help to address the pathological systems of these diseases. This review presents our understanding of the potential mechanism of PANoptosis and the role of PANoptosis in different pulmonary diseases.
Collapse
Affiliation(s)
| | | | | | - Longshuang Huang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (S.C.); (J.J.); (T.L.)
| |
Collapse
|
17
|
Davis D, Thadhani J, Choudhary V, Nausheem R, Vallejo-Zambrano CR, Mohammad Arifuddin B, Ali M, Carson BJ, Kanwal F, Nagarajan L. Advancements in the Management of Severe Community-Acquired Pneumonia: A Comprehensive Narrative Review. Cureus 2023; 15:e46893. [PMID: 37954793 PMCID: PMC10638673 DOI: 10.7759/cureus.46893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2023] [Indexed: 11/14/2023] Open
Abstract
Pneumonia, classified as a lower respiratory tract illness, affects different parts of the bronchial system as well as alveoli and can present with varying severities depending on co-morbidities and causative pathogens. It can be broadly classified using the setting in which it was acquired, namely the community or hospital setting, the former being more common and spreading through person-to-person droplet transmission. Community-acquired pneumonia (CAP) is currently the fourth leading cause of death worldwide, and its high mortality makes continual insight into the management of the condition worthwhile. This review explores the literature specifically for severe CAP (sCAP) and delves into the diagnosis, various modalities of treatment, and management of the condition. This condition can be defined as pneumonia requiring mechanical ventilation in the ICU and/or presenting with sepsis and organ failure due to pneumonia. The disease process is characterized by inflammation of the lung parenchyma, initiated by a combination of pathogens and lowered local defenses. Acute diagnosis of the condition is vital in reducing negative patient outcomes, namely through clinical presentation, blood/sputum cultures, imaging modalities such as computed tomography scan, and inflammatory markers, identifying common causative pathogens such as Streptococcus pneumoniae, rhinovirus, Legionella, and viral influenza. Pathogens such as Escherichia coli should also be investigated in patients with chronic obstructive pulmonary disease. The mainstay of treating sCAP includes rapid ICU admission once a diagnosis has been confirmed, initiating sepsis protocol, and treatment with combined empiric antibiotic regimens consisting of beta-lactams and macrolides. Corticosteroid use alongside antibiotics shows promise in reducing inflammation, but its use has to be judged on a case-by-case basis. New drugs such as omadacycline, delafloxacin, and zabofloxacin have shown valid evidence for the treatment of resistant causative organisms. The main guidelines for preventing sCAP include maintaining a healthy lifestyle, and annual pneumococcal and influenza vaccines are recommended for the most vulnerable patient groups, such as those with COPD and immunosuppression.
Collapse
Affiliation(s)
- Don Davis
- Medicine, Medical University of Varna, Varna, BGR
| | - Jainisha Thadhani
- Medicine, Royal College of Surgeons in Ireland, Medical University of Bahrain, Manama, BHR
| | | | | | | | | | - Mujahaith Ali
- Medicine, Ternopil National Medical University, Ternopil, UKR
| | - Bryan J Carson
- Emergency Medicine, Northern Health and Social Care Trust, Coleraine, GBR
| | - Fnu Kanwal
- Medical College, Chandka Medical College, Larkana, PAK
| | - Lavanya Nagarajan
- Department of Medicine, The Tamilnadu Dr.M.G.R. Medical University, Chennai, IND
| |
Collapse
|
18
|
Bazant J, Ott B, Hudel M, Hain T, Lucas R, Mraheil MA. Impact of Endogenous Pneumococcal Hydrogen Peroxide on the Activity and Release of Pneumolysin. Toxins (Basel) 2023; 15:593. [PMID: 37888624 PMCID: PMC10611280 DOI: 10.3390/toxins15100593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/21/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
Streptococcus pneumoniae is the leading cause of community-acquired pneumonia. The pore-forming cholesterol-dependent cytolysin (CDC) pneumolysin (PLY) and the physiological metabolite hydrogen peroxide (H2O2) can greatly increase the virulence of pneumococci. Although most studies have focused on the contribution of both virulence factors to the course of pneumococcal infection, it is unknown whether or how H2O2 can affect PLY activity. Of note, S. pneumoniae exploits endogenous H2O2 as an intracellular signalling molecule to modulate the activity of several proteins. Here, we demonstrate that H2O2 negatively affects the haemolytic activity of PLY in a concentration-dependent manner. Prevention of cysteine-dependent sulfenylation upon substitution of the unique and highly conserved cysteine residue to serine in PLY significantly reduces the toxin's susceptibility to H2O2 treatment and completely abolishes the ability of DTT to activate PLY. We also detect a clear gradual correlation between endogenous H2O2 generation and PLY release, with decreased H2O2 production causing a decline in the release of PLY. Comparative transcriptome sequencing analysis of the wild-type S. pneumoniae strain and three mutants impaired in H2O2 production indicates enhanced expression of several genes involved in peptidoglycan (PG) synthesis and in the production of choline-binding proteins (CPBs). One explanation for the impact of H2O2 on PLY release is the observed upregulation of the PG bridge formation alanyltransferases MurM and MurN, which evidentially negatively affect the PLY release. Our findings shed light on the significance of endogenous pneumococcal H2O2 in controlling PLY activity and release.
Collapse
Affiliation(s)
- Jasmin Bazant
- Institute of Medical Microbiology, German Center for Infection Research, Partner Site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Schubertstrasse 81, 35392 Giessen, Germany; (J.B.); (B.O.); (M.H.); (T.H.)
| | - Benjamin Ott
- Institute of Medical Microbiology, German Center for Infection Research, Partner Site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Schubertstrasse 81, 35392 Giessen, Germany; (J.B.); (B.O.); (M.H.); (T.H.)
| | - Martina Hudel
- Institute of Medical Microbiology, German Center for Infection Research, Partner Site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Schubertstrasse 81, 35392 Giessen, Germany; (J.B.); (B.O.); (M.H.); (T.H.)
| | - Torsten Hain
- Institute of Medical Microbiology, German Center for Infection Research, Partner Site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Schubertstrasse 81, 35392 Giessen, Germany; (J.B.); (B.O.); (M.H.); (T.H.)
| | - Rudolf Lucas
- Vascular Biology Center, Department of Pharmacology and Toxicology and Division of Pulmonary Critical Care Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA;
| | - Mobarak Abu Mraheil
- Institute of Medical Microbiology, German Center for Infection Research, Partner Site Giessen-Marburg-Langen, Justus-Liebig University Giessen, Schubertstrasse 81, 35392 Giessen, Germany; (J.B.); (B.O.); (M.H.); (T.H.)
| |
Collapse
|
19
|
Sharma S, Pellett S, Morse SA. Special Issue: Gram-Positive Bacterial Toxins. Microorganisms 2023; 11:2054. [PMID: 37630614 PMCID: PMC10459523 DOI: 10.3390/microorganisms11082054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
The Gram stain classifies most bacteria into one of two groups, Gram-negative or Gram-positive, based on the composition of their cell walls [...].
Collapse
Affiliation(s)
- Shashi Sharma
- Division of Microbiology, Office of Regulatory Science, CFSAN/US Food and Drug Administration, College Park, MD 20740, USA
| | - Sabine Pellett
- Department of Bacteriology, University of Wisconsin, 1550 Linden Drive, Madison, WI 53706, USA
| | - Stephen A Morse
- IHRC, Inc., 2 Ravinia Drive, Suite 1200, Atlanta, GA 30346, USA
| |
Collapse
|
20
|
Xu J, Xie L. Advances in immune response to pulmonary infection: Nonspecificity, specificity and memory. Chronic Dis Transl Med 2023; 9:71-81. [PMID: 37305110 PMCID: PMC10249196 DOI: 10.1002/cdt3.71] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 04/02/2023] [Accepted: 04/14/2023] [Indexed: 06/13/2023] Open
Abstract
The lung immune response consists of various cells involved in both innate and adaptive immune processes. Innate immunity participates in immune resistance in a nonspecific manner, whereas adaptive immunity effectively eliminates pathogens through specific recognition. It was previously believed that adaptive immune memory plays a leading role during secondary infections; however, innate immunity is also involved in immune memory. Trained immunity refers to the long-term functional reprogramming of innate immune cells caused by the first infection, which alters the immune response during the second challenge. Tissue resilience limits the tissue damage caused by infection by controlling excessive inflammation and promoting tissue repair. In this review, we summarize the impact of host immunity on the pathophysiological processes of pulmonary infections and discuss the latest progress in this regard. In addition to the factors influencing pathogenic microorganisms, we emphasize the importance of the host response.
Collapse
Affiliation(s)
- Jianqiao Xu
- College of Pulmonary & Critical Care Medicine, 8th Medical CenterChinese PLA General HospitalBeijingChina
- Medical School of Chinese PLABeijingChina
| | - Lixin Xie
- College of Pulmonary & Critical Care Medicine, 8th Medical CenterChinese PLA General HospitalBeijingChina
- Medical School of Chinese PLABeijingChina
| |
Collapse
|
21
|
Kouakou YI, Lee RJ. Interkingdom Detection of Bacterial Quorum-Sensing Molecules by Mammalian Taste Receptors. Microorganisms 2023; 11:1295. [PMID: 37317269 PMCID: PMC10221136 DOI: 10.3390/microorganisms11051295] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 06/16/2023] Open
Abstract
Bitter and sweet taste G protein-coupled receptors (known as T2Rs and T1Rs, respectively) were originally identified in type II taste cells on the tongue, where they signal perception of bitter and sweet tastes, respectively. Over the past ~15 years, taste receptors have been identified in cells all over the body, demonstrating a more general chemosensory role beyond taste. Bitter and sweet taste receptors regulate gut epithelial function, pancreatic β cell secretion, thyroid hormone secretion, adipocyte function, and many other processes. Emerging data from a variety of tissues suggest that taste receptors are also used by mammalian cells to "eavesdrop" on bacterial communications. These receptors are activated by several quorum-sensing molecules, including acyl-homoserine lactones and quinolones from Gram-negative bacteria such as Pseudomonas aeruginosa, competence stimulating peptides from Streptococcus mutans, and D-amino acids from Staphylococcus aureus. Taste receptors are an arm of immune surveillance similar to Toll-like receptors and other pattern recognition receptors. Because they are activated by quorum-sensing molecules, taste receptors report information about microbial population density based on the chemical composition of the extracellular environment. This review summarizes current knowledge of bacterial activation of taste receptors and identifies important questions remaining in this field.
Collapse
Affiliation(s)
- Yobouet Ines Kouakou
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Robert J. Lee
- Department of Otorhinolaryngology and Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
22
|
PLGA-Based Micro/Nanoparticles: An Overview of Their Applications in Respiratory Diseases. Int J Mol Sci 2023; 24:ijms24054333. [PMID: 36901762 PMCID: PMC10002081 DOI: 10.3390/ijms24054333] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/24/2023] Open
Abstract
Respiratory diseases, such as asthma and chronic obstructive pulmonary disease (COPD), are critical areas of medical research, as millions of people are affected worldwide. In fact, more than 9 million deaths worldwide were associated with respiratory diseases in 2016, equivalent to 15% of global deaths, and the prevalence is increasing every year as the population ages. Due to inadequate treatment options, the treatments for many respiratory diseases are limited to relieving symptoms rather than curing the disease. Therefore, new therapeutic strategies for respiratory diseases are urgently needed. Poly (lactic-co-glycolic acid) micro/nanoparticles (PLGA M/NPs) have good biocompatibility, biodegradability and unique physical and chemical properties, making them one of the most popular and effective drug delivery polymers. In this review, we summarized the synthesis and modification methods of PLGA M/NPs and their applications in the treatment of respiratory diseases (asthma, COPD, cystic fibrosis (CF), etc.) and also discussed the research progress and current research status of PLGA M/NPs in respiratory diseases. It was concluded that PLGA M/NPs are the promising drug delivery vehicles for the treatment of respiratory diseases due to their advantages of low toxicity, high bioavailability, high drug loading capacity, plasticity and modifiability. And at the end, we presented an outlook on future research directions, aiming to provide some new ideas for future research directions and hopefully to promote their widespread application in clinical treatment.
Collapse
|
23
|
Therapeutic potential of kaempferol on Streptococcus pneumoniae infection. Microbes Infect 2023; 25:105058. [PMID: 36216303 PMCID: PMC9540706 DOI: 10.1016/j.micinf.2022.105058] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 11/07/2022]
Abstract
Co-infections with pathogens and secondary bacterial infections play significant roles during the pandemic coronavirus disease 2019 (COVID-19) pathogenetic process, caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Notably, co-infections with Streptococcus pneumoniae (S. pneumoniae), as a major Gram-positive pathogen causing pneumonia or meningitis, severely threaten the diagnosis, therapy, and prognosis of COVID-19 worldwide. Accumulating evidences have emerged indicating that S. pneumoniae evolves multiple virulence factors, including pneumolysin (PLY) and sortase A (SrtA), which have been extensively explored as alternative anti-infection targets. In our study, natural flavonoid kaempferol was identified as a potential candidate drug for infection therapeutics via anti-virulence mechanisms. We found that kaempferol could interfere with the pore-forming activity of PLY by engaging with catalytic active sites and consequently inhibit PLY-mediated cytotoxicity. Additionally, exposed to kaempferol significantly reduced the SrtA peptidase activity by occupying the active sites of SrtA. Further, the biofilms formation and bacterial adhesion to the host cells could be significantly thwarted by kaempferol incubation. In vivo infection model by S. pneumoniae highlighted that kaempferol oral administration exhibited notable treatment benefits, as evidenced by decreased bacterial burden, suggesting that kaempferol has tremendous potential to attenuate S. pneumoniae pathogenicity. Scientifically, our study implies that kaempferol is a promising therapeutic option by targeting bacterial virulence factors.
Collapse
|
24
|
Xie S, Wang J, Tuo W, Zhuang S, Cai Q, Yao C, Han F, Zhu H, Xiang Y, Yuan C. Serum level of S100A8/A9 as a biomarker for establishing the diagnosis and severity of community-acquired pneumonia in children. Front Cell Infect Microbiol 2023; 13:1139556. [PMID: 37180431 PMCID: PMC10172663 DOI: 10.3389/fcimb.2023.1139556] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 04/13/2023] [Indexed: 05/16/2023] Open
Abstract
Background S100A8/A9, which is a member of S100 proteins, may be involved in the pathophysiology of Community-acquired pneumonia (CAP) that seriously threatens children's health. However, circulating markers to assess the severity of pneumonia in children are yet to be explored. Therefore, we aimed to investigate the diagnostic performance of serum S100A8/A9 level in determining the severity of CAP in children. Methods In this prospective and observational study, we recruited 195 in-hospital children diagnosed with CAP. In comparison, 63 healthy children (HC) and 58 children with non-infectious pneumonia (pneumonitis) were included as control groups. Demographic and clinical data were collected. Serum S100A8/A9 levels, serum pro-calcitonin concentrations, and blood leucocyte counts were quantified. Results The serum S100A8/A9 levels in patients with CAP was 1.59 ± 1.32 ng/mL, which was approximately five and two times higher than those in healthy controls and those in children with pneumonitis, respectively. Serum S100A8/A9 was elevated parallelly with the clinical pulmonary infection score. The sensitivity, specificity, and Youden's index of S100A8/A9 ≥1.25 ng/mL for predicting the severity of CAP in children was optimal. The area under the receiver operating characteristic curve of S100A8/A9 was the highest among the indices used to evaluate severity. Conclusions S100A8/A9 may serve as a biomarker for predicting the severity of the condition in children with CAP and establishing treatment grading.
Collapse
Affiliation(s)
- Si Xie
- Department of Laboratory Medicine, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Wang
- Department of Laboratory Medicine, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenbin Tuo
- Department of Laboratory Medicine, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shihao Zhuang
- Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Qinzhen Cai
- Department of Laboratory Medicine, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong Yao
- Health Care Department, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Han
- Department of Pediatric Respiratory Medicine, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongmin Zhu
- Department of Neurology, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Hongmin Zhu, ; Yun Xiang, ; Chunhui Yuan,
| | - Yun Xiang
- Department of Laboratory Medicine, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Hongmin Zhu, ; Yun Xiang, ; Chunhui Yuan,
| | - Chunhui Yuan
- Department of Laboratory Medicine, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Hongmin Zhu, ; Yun Xiang, ; Chunhui Yuan,
| |
Collapse
|
25
|
Langouët-Astrié C, Oshima K, McMurtry SA, Yang Y, Kwiecinski JM, LaRivière WB, Kavanaugh JS, Zakharevich I, Hansen KC, Shi D, Zhang F, Boguslawski KM, Perelman SS, Su G, Torres VJ, Liu J, Horswill AR, Schmidt EP. The influenza-injured lung microenvironment promotes MRSA virulence, contributing to severe secondary bacterial pneumonia. Cell Rep 2022; 41:111721. [PMID: 36450248 PMCID: PMC10082619 DOI: 10.1016/j.celrep.2022.111721] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 10/12/2022] [Accepted: 11/03/2022] [Indexed: 12/03/2022] Open
Abstract
Influenza infection is substantially worsened by the onset of secondary pneumonia caused by bacteria, such as methicillin-resistant Staphylococcus aureus (MRSA). The bidirectional interaction between the influenza-injured lung microenvironment and MRSA is poorly understood. By conditioning MRSA ex vivo in bronchoalveolar lavage fluid collected from mice at various time points of influenza infection, we found that the influenza-injured lung microenvironment dynamically induces MRSA to increase cytotoxin expression while decreasing metabolic pathways. LukAB, a SaeRS two-component system-dependent cytotoxin, is particularly important to the severity of post-influenza MRSA pneumonia. LukAB's activity is likely shaped by the post-influenza lung microenvironment, as LukAB binds to (and is activated by) heparan sulfate (HS) oligosaccharide sequences shed from the epithelial glycocalyx after influenza. Our findings indicate that post-influenza MRSA pneumonia is shaped by bidirectional host-pathogen interactions: host injury triggers changes in bacterial expression of toxins, the activity of which may be shaped by host-derived HS fragments.
Collapse
Affiliation(s)
| | - Kaori Oshima
- Division of Pulmonary Sciences and Critical Care, University of Colorado Denver, Aurora, CO 80045, USA
| | - Sarah A McMurtry
- Division of Pulmonary Sciences and Critical Care, University of Colorado Denver, Aurora, CO 80045, USA
| | - Yimu Yang
- Division of Pulmonary Sciences and Critical Care, University of Colorado Denver, Aurora, CO 80045, USA
| | - Jakub M Kwiecinski
- Department of Immunology & Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA; Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow 30387, Poland
| | - Wells B LaRivière
- Division of Pulmonary Sciences and Critical Care, University of Colorado Denver, Aurora, CO 80045, USA; Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jeffrey S Kavanaugh
- Department of Immunology & Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Igor Zakharevich
- Department of Biochemistry and Molecular Genetics, University of Colorado, Aurora, CO 80045, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado, Aurora, CO 80045, USA
| | - Deling Shi
- Department of Chemistry, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Fuming Zhang
- Department of Chemistry, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Kristina M Boguslawski
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Sofya S Perelman
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Gouwei Su
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | - Victor J Torres
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Jian Liu
- University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | - Alexander R Horswill
- Department of Immunology & Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Eric P Schmidt
- Division of Pulmonary Sciences and Critical Care, University of Colorado Denver, Aurora, CO 80045, USA; Department of Medicine, Massachusetts General Hospital, Boston, MA 02115, USA
| |
Collapse
|
26
|
Ciptaningtyas VR, Hapsari R, Lestari ES, Farida H, de Mast Q, de Jonge MI. Bacterial colonization of the upper airways of children positive and negative for SARS-CoV-2 during the COVID-19 pandemic. BMC Infect Dis 2022; 22:860. [PMID: 36396997 PMCID: PMC9670079 DOI: 10.1186/s12879-022-07851-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 11/08/2022] [Indexed: 11/18/2022] Open
Abstract
Background Our understanding of the influence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection on bacterial colonization in the children’s upper nasopharyngeal tract during the coronavirus infectious disease (COVID-19) pandemic is limited. This study aimed to determine whether there were any differences in bacterial colonization between asymptomatic children with or without a positive SARS-CoV-2 quantitative reverse transcriptase-polymerase chain reaction (RT-qPCR) results in the community setting. Methods A cross-sectional community-based exploratory study was conducted from March to May 2021 in Semarang, Central Java Province, Indonesia. Using stored nasopharyngeal swabs collected from children under 18 years as a contact tracing program, we performed a real-time quantitative (qPCR) for the most important bacterial colonizing pathogens: Streptococcus pneumoniae, Haemophilus influenzae, Staphylococcus aureus, and Klebsiella pneumoniae. Results Swabs from a total of 440 children were included in this study, of which 228 (51.8%) were RT-qPCR-confirmed SARS-CoV-2 positive. In the 440 children, colonization rates were highest for H. influenzae (61.4%), followed by S. pneumoniae (17.5%), S. aureus (12.0%), and K. pneumoniae (1.8%). The co-occurrence of both S. pneumoniae and H. influenzae in the upper respiratory tract was significantly associated with a SARS-CoV-2 negative RT-qPCR. In contrast, colonization with only S. aureus was more common in SARS-CoV-2-positive children. Conclusion Overall, this exploratory study concludes that there is a significant difference in the bacterial nasopharyngeal colonization pattern between SARS-CoV-2 positive and negative in asymptomatic children in the community in Indonesia. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-022-07851-z.
Collapse
|
27
|
Grousd JA, Dresden BP, Riesmeyer AM, Cooper VS, Bomberger JM, Richardson AR, Alcorn JF. Novel Requirement for Staphylococcal Cell Wall-Anchored Protein SasD in Pulmonary Infection. Microbiol Spectr 2022; 10:e0164522. [PMID: 36040164 PMCID: PMC9603976 DOI: 10.1128/spectrum.01645-22] [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: 07/15/2022] [Accepted: 08/16/2022] [Indexed: 02/04/2023] Open
Abstract
Staphylococcus aureus can complicate preceding viral infections, including influenza virus. A bacterial infection combined with a preceding viral infection, known as superinfection, leads to worse outcomes than a single infection. Most of the pulmonary infection literature focuses on the changes in immune responses to bacteria between homeostatic and virally infected lungs. However, it is unclear how much of an influence bacterial virulence factors have in single or superinfection. Staphylococcal species express a broad range of cell wall-anchored proteins (CWAs) that have roles in host adhesion, nutrient acquisition, and immune evasion. We screened the importance of these CWAs using mutants lacking individual CWAs in vivo in both bacterial pneumonia and influenza superinfection. In bacterial pneumonia, the lack of individual CWAs leads to various decreases in bacterial burden, lung damage, and immune infiltration into the lung. However, the presence of a preceding influenza infection partially abrogates the requirement for CWAs. In the screen, we found that the uncharacterized CWA S. aureus surface protein D (SasD) induced changes in both inflammatory and homeostatic lung markers. We further characterized a SasD mutant (sasD A50.1) in the context of pneumonia. Mice infected with sasD A50.1 have decreased bacterial burden, inflammatory responses, and mortality compared to wild-type S. aureus. Mice also have reduced levels of interleukin-1β (IL-1β), likely derived from macrophages. Reductions in IL-1β transcript levels as well as increased macrophage viability point at differences in cell death pathways. These data identify a novel virulence factor for S. aureus that influences inflammatory signaling within the lung. IMPORTANCE Staphylococcus aureus is a common commensal bacterium that can cause severe infections, such as pneumonia. In the lung, viral infections increase the risk of staphylococcal pneumonia, leading to combined infections known as superinfections. The most common virus associated with S. aureus pneumonia is influenza, and superinfections lead to worse patient outcomes than either infection alone. While there is much known about how the immune system differs between healthy and virally infected lungs, the role of bacterial virulence factors in single and superinfection is less understood. The significance of our research is identifying bacterial components that play a role in the initiation of lung injury, which could lead to future therapies to prevent pulmonary single or superinfection with S. aureus.
Collapse
Affiliation(s)
- Jennifer A. Grousd
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Brooke P. Dresden
- Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Abigail M. Riesmeyer
- Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Vaughn S. Cooper
- Department of Microbiology & Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jennifer M. Bomberger
- Department of Microbiology & Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anthony R. Richardson
- Department of Microbiology & Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - John F. Alcorn
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
28
|
Li E, Yan R, Yan K, Zhang R, Zhang Q, Zou P, Wang H, Qiao H, Li S, Ma Q, Liao B. Single-cell RNA sequencing reveals the role of immune-related autophagy in spinal cord injury in rats. Front Immunol 2022; 13:987344. [PMID: 36211348 PMCID: PMC9535363 DOI: 10.3389/fimmu.2022.987344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
Spinal cord injury refers to damage to the spinal cord due to trauma, disease, or degeneration; and the number of new cases is increasing yearly. Significant cellular changes are known to occur in the area of spinal cord injury. However, changes in cellular composition, trajectory of cell development, and intercellular communication in the injured area remain unclear. Here, we used single-cell RNA sequencing to evaluate almost all the cell types that constitute the site of spinal cord injury in rats. In addition to mapping the cells of the injured area, we screened the expression of immune autophagy-related factors in cells and identified signaling pathways by the measuring the expression of the receptor−ligand pairs to regulate specific cell interactions during autophagy after spinal cord injury. Our data set is a valuable resource that provides new insights into the pathobiology of spinal cord injury and other traumatic diseases of the central nervous system.
Collapse
Affiliation(s)
- Erliang Li
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi’an, China
| | - Rongbao Yan
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kang Yan
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi’an, China
| | - Rui Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi’an, China
| | - Qian Zhang
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi’an, China
| | - Peng Zou
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi’an, China
| | - Huimei Wang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huan Qiao
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi’an, China
| | - Shuang Li
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi’an, China
| | - Qiong Ma
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi’an, China
- *Correspondence: Bo Liao, ; Qiong Ma,
| | - Bo Liao
- Department of Orthopaedics, The Second Affiliated Hospital of Air Force Military Medical University, Xi’an, China
- *Correspondence: Bo Liao, ; Qiong Ma,
| |
Collapse
|
29
|
The pro-inflammatory effect of Staphylokinase contributes to community-associated Staphylococcus aureus pneumonia. Commun Biol 2022; 5:618. [PMID: 35739262 PMCID: PMC9226170 DOI: 10.1038/s42003-022-03571-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 06/09/2022] [Indexed: 11/09/2022] Open
Abstract
Pneumonia caused by community-associated Staphylococcus aureus (CA-SA) has high morbidity and mortality, but its pathogenic mechanism remains to be further investigated. Herein, we identify that staphylokinase (SAK) is significantly induced in CA-SA and inhibits biofilm formation in a plasminogen-dependent manner. Importantly, SAK can enhance CA-SA-mediated pneumonia in both wild-type and cathelicidins-related antimicrobial peptide knockout (CRAMP−/−) mice, suggesting that SAK exacerbates pneumonia in a CRAMP-independent manner. Mechanistically, SAK induces pro-inflammatory effects, especially in the priming step of NLRP3 inflammasome activation. Moreover, we demonstrate that SAK can increase K+ efflux, production of reactive oxygen species production, and activation of NF-κB signaling. Furthermore, the NLRP3 inflammasome inhibitor can counteract the effective of SAK induced CA-SA lung infection in mice. Taken together, we speculate that SAK exacerbates CA-SA-induced pneumonia by promoting NLRP3 inflammasome activation, providing new insights into the pathogenesis of highly virulent CA-SA and emphasizes the importance of controlling inflammation in acute pneumonia. Staphylokinase (Sak) is highly prevalent in human-adapted S. aureus strains, with increased expression in community-associated (CA-SA) strains, promoting lung infection and activation of the NLRP3 inflammasome.
Collapse
|
30
|
Li Z, Liu Y, Zhang L. Role of the microbiome in oral cancer occurrence, progression and therapy. Microb Pathog 2022; 169:105638. [PMID: 35718272 DOI: 10.1016/j.micpath.2022.105638] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 06/07/2022] [Accepted: 06/10/2022] [Indexed: 02/07/2023]
Abstract
The oral cavity, like other digestive or mucosal sites, contains a site-specific microbiome that plays a significant role in maintaining health and homeostasis. Strictly speaking, the gastrointestinal tract starts from the oral cavity, with special attention paid to the specific flora of the oral cavity. In healthy people, the microbiome of the oral microenvironment is governed by beneficial bacteria, that benefit the host by symbiosis. When a microecological imbalance occurs, changes in immune and metabolic signals affect the characteristics of cancer, as well as chronic inflammation, disruption of the epithelial barrier, changes in cell proliferation and cell apoptosis, genomic instability, angiogenesis, and epithelial barrier destruction and metabolic regulation. These pathophysiological changes could result in oral cancer. Rising evidence suggests that oral dysbacteriosis and particular microbes may play a positive role in the evolution, development, progression, and metastasis of oral cancer, for instance, oral squamous cell carcinoma (OSCC) through direct or indirect action.
Collapse
Affiliation(s)
- Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000, China.
| | - Yuan Liu
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000, China.
| | - Ling Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000, China.
| |
Collapse
|
31
|
Okahashi N, Sumitomo T, Nakata M, Kawabata S. Secondary streptococcal infection following influenza. Microbiol Immunol 2022; 66:253-263. [PMID: 35088451 DOI: 10.1111/1348-0421.12965] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/18/2022] [Accepted: 01/24/2022] [Indexed: 12/01/2022]
Abstract
Secondary bacterial infection following influenza A virus (IAV) infection is a major cause of morbidity and mortality during influenza epidemics. Streptococcus pneumoniae has been identified as a predominant pathogen in secondary pneumonia cases that develop following influenza. Although IAV has been shown to enhance susceptibility to the secondary bacterial infection, the underlying mechanism of the viral-bacterial synergy leading to disease progression is complex and remains elusive. In this review, cooperative interactions of viruses and streptococci during co- or secondary infection with IAV are described. IAV infects the upper respiratory tract, therefore, streptococci that inhabit or infect the respiratory tract are of special interest. Since many excellent reviews on the co-infection of IAV and S. pneumoniae have already been published, this review is intended to describe the unique interactions between other streptococci and IAV. Both streptococcal and IAV infections modulate the host epithelial barrier of the respiratory tract in various ways. IAV infection directly disrupts epithelial barriers, though at the same time the virus modifies the properties of infected cells to enhance streptococcal adherence and invasion. Mitis group streptococci produce neuraminidases, which promote IAV infection in a unique manner. The studies reviewed here have revealed intriguing mechanisms underlying secondary streptococcal infection following influenza. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Nobuo Okahashi
- Center for Frontier Oral Science, Osaka University Graduate School of Dentistry, Suita-Osaka, Japan
| | - Tomoko Sumitomo
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Suita-Osaka, Japan
| | - Masanobu Nakata
- Department of Oral Microbiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Shigetada Kawabata
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Suita-Osaka, Japan
| |
Collapse
|
32
|
Xie F, Chen R, Zhao J, Xu C, Zan C, Yue B, Tian W, Yi W. Cell cycle kinase CHEK2 in macrophages alleviates the inflammatory response to Staphylococcus aureus-induced pneumonia. Exp Lung Res 2022; 48:53-60. [PMID: 35075953 DOI: 10.1080/01902148.2022.2029625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Excessive macrophage-mediated inflammation participates in the development of Staphylococcus aureus (S. aureus)-induced pneumonia. Checkpoint kinase 2 (Chek2) was screened out as macrophage-related infantile pneumonia gene after the differentially expressed analysis of RNAseq data derived from pam3CSK4 stimulated bone marrow-derived macrophages (BMDMs). METHODS RAW264.7 macrophage cells were transfected with Chek2-specific gRNA, which were further overexpressed with wide-type Chek2 or Chek2 kinase activity mutant (Chek2 KD, D368N). At the same time, the relative protein and mRNA expression of inflammatory cytokines were determined. C57BL/6J WT mice were intranasally infected with S. aureus to induce S. aureus-induced pneumonia, which was treated with BML-277, an inhibitor of Chek2. The symptoms of pneumonia mice and inflammatory cytokines associated with the nuclear factor kappa B (NF-κB) signaling pathways were further examined. RESULTS In vivo, BML-277 significantly promoted pneumonia symptoms, including mortality, lung infiltration of immune cells, and the abundance of lung pro-inflammatory cytokines. Mechanically, BML-277 did not affect BMDMs survival but up-regulated the mRNA expression of tumor necrosis factor (Tnf), nitric oxide synthase 2 (Nos2), interleukin (Il)23a, and the secretion of Tnf-α and Il-23a. At the same time, genetic complementation experiment testified that Chek2 KD did not inhibit NF-κB and relevant inflammatory cytokines expression. CONCLUSION Chek2 functions through the kinase mechanism to down-regulate the NF-κB pathway in macrophages to alleviate S. aureus-induced pneumonia in mice.
Collapse
Affiliation(s)
- Fei Xie
- Department of Pediatrics, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Ruidong Chen
- Department of Pediatrics, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Jie Zhao
- Department of Pediatrics, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Chunyan Xu
- Department of Pediatrics, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Chunxiang Zan
- Child Healthcare Department, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Bin Yue
- Department of Pediatrics, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Wenqiu Tian
- Department of Pediatrics, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Wenxia Yi
- Department of Pediatrics, Cangzhou Central Hospital, Cangzhou, Hebei, China
| |
Collapse
|
33
|
Qu J, Liu Q, You G, Ye L, Jin Y, Kong L, Guo W, Xu Q, Sun Y. Advances in ameliorating inflammatory diseases and cancers by andrographolide: Pharmacokinetics, pharmacodynamics, and perspective. Med Res Rev 2021; 42:1147-1178. [PMID: 34877672 DOI: 10.1002/med.21873] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/07/2021] [Accepted: 11/10/2021] [Indexed: 12/26/2022]
Abstract
Andrographolide, a well-known natural lactone having a range of pharmacological actions in traditional Chinese medicine. It has long been used to cure a variety of ailments. In this review, we cover the pharmacokinetics and pharmacological activity of andrographolide which supports its further clinical application in cancers and inflammatory diseases. Growing evidence shows a good therapeutic effect in inflammatory diseases, including liver diseases, joint diseases, respiratory system diseases, nervous system diseases, heart diseases, inflammatory bowel diseases, and inflammatory skin diseases. As a result, the effects of andrographolide on immune cells and the processes that underpin them are discussed. The preclinical use of andrographolide to different organs in response to malignancies such as colorectal, liver, gastric, breast, prostate, lung, and oral cancers has also been reviewed. In addition, several clinical trials of andrographolide in inflammatory diseases and cancers have been summarized. This review highlights recent advances in ameliorating inflammatory diseases as well as cancers by andrographolide and its analogs, providing a new perspective for subsequent research of this traditional natural product.
Collapse
Affiliation(s)
- Jiao Qu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, Nanjing, China
| | - Qianqian Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, Nanjing, China
| | - Guoquan You
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, Nanjing, China
| | - Ling Ye
- Biopharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Lingdong Kong
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, Nanjing, China
| | - Wenjie Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, Nanjing, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, Nanjing, China.,Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Biotechnology and Pharmaceutical Sciences, School of Life Science, Nanjing University, Nanjing, China.,Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| |
Collapse
|
34
|
Su R, Zhang Y, Zhang J, Wang H, Luo Y, Chan HF, Tao Y, Chen Z, Li M. Nanomedicine to advance the treatment of bacteria-induced acute lung injury. J Mater Chem B 2021; 9:9100-9115. [PMID: 34672317 DOI: 10.1039/d1tb01770e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bacteria-induced acute lung injury (ALI) is associated with a high mortality rate due to the lack of an effective treatment. Patients often rely on supportive care such as low tidal volume ventilation to alleviate the symptoms. Nanomedicine has recently received much attention owing to its premium benefits of delivering drugs in a sustainable and controllable manner while minimizing the potential side effects. It can effectively improve the prognosis of bacteria-induced ALI through targeted delivery of drugs, regulation of multiple inflammatory pathways, and combating antibiotic resistance. Hence, in this review, we first discuss the pathogenesis of ALI and its potential therapeutics. In particular, the state-of-the-art nanomedicines for the treatment of bacteria-induced ALI are highlighted, including their administration routes, in vivo distribution, and clearance. Furthermore, the available bacteria-induced ALI animal models are also summarized. In the end, future perspectives of nanomedicine for ALI treatment are proposed.
Collapse
Affiliation(s)
- Ruonan Su
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yu Zhang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca 14853, USA
| | - Jiabin Zhang
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Haixia Wang
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yun Luo
- Department of Urology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Science, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Yu Tao
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Zhuanggui Chen
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Mingqiang Li
- Center for Nanomedicine, Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China. .,Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, China
| |
Collapse
|
35
|
Chen H, Yin Y, van Dorp L, Shaw LP, Gao H, Acman M, Yuan J, Chen F, Sun S, Wang X, Li S, Zhang Y, Farrer RA, Wang H, Balloux F. Drivers of methicillin-resistant Staphylococcus aureus (MRSA) lineage replacement in China. Genome Med 2021; 13:171. [PMID: 34711267 PMCID: PMC8555231 DOI: 10.1186/s13073-021-00992-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 10/17/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Methicillin-resistant Staphylococcus aureus (MRSA) is a major nosocomial pathogen subdivided into lineages termed sequence types (STs). Since the 1950s, successive waves of STs have appeared and replaced previously dominant lineages. One such event has been occurring in China since 2013, with community-associated (CA-MRSA) strains including ST59 largely replacing the previously dominant healthcare-associated (HA-MRSA) ST239. We previously showed that ST59 isolates tend to have a competitive advantage in growth experiments against ST239. However, the underlying genomic and phenotypic drivers of this replacement event are unclear. METHODS Here, we investigated the replacement of ST239 using whole-genome sequencing data from 204 ST239 and ST59 isolates collected in Chinese hospitals between 1994 and 2016. We reconstructed the evolutionary history of each ST and considered two non-mutually exclusive hypotheses for ST59 replacing ST239: antimicrobial resistance (AMR) profile and/or ability to colonise and persist in the environment through biofilm formation. We also investigated the differences in cytolytic activity, linked to higher virulence, between STs. We performed an association study using the presence and absence of accessory virulence genes. RESULTS ST59 isolates carried fewer AMR genes than ST239 and showed no evidence of evolving towards higher AMR. Biofilm production was marginally higher in ST59 overall, though this effect was not consistent across sub-lineages so is unlikely to be a sole driver of replacement. Consistent with previous observations of higher virulence in CA-MRSA STs, we observed that ST59 isolates exhibit significantly higher cytolytic activity than ST239 isolates, despite carrying on average fewer putative virulence genes. Our association study identified the chemotaxis inhibitory protein (chp) as a strong candidate for involvement in the increased virulence potential of ST59. We experimentally validated the role of chp in increasing the virulence potential of ST59 by creating Δchp knockout mutants, confirming that ST59 can carry chp without a measurable impact on fitness. CONCLUSIONS Our results suggest that the ongoing replacement of ST239 by ST59 in China is not associated to higher AMR carriage or biofilm production. However, the increase in ST59 prevalence is concerning since it is linked to a higher potential for virulence, aided by the carriage of the chp gene.
Collapse
Affiliation(s)
- Hongbin Chen
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, 100044, China
- UCL Genetics Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Yuyao Yin
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, 100044, China
| | - Lucy van Dorp
- UCL Genetics Institute, University College London, Gower Street, London, WC1E 6BT, UK.
| | - Liam P Shaw
- UCL Genetics Institute, University College London, Gower Street, London, WC1E 6BT, UK
- Department of Zoology, University of Oxford, Oxford, OX1 3SZ, UK
| | - Hua Gao
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, 100044, China
| | - Mislav Acman
- UCL Genetics Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Jizhen Yuan
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, 100044, China
- The No. 971 Hospital of People's Liberation Army Navy, Qingdao, 266000, Shandong, China
| | - Fengning Chen
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, 100044, China
| | - Shijun Sun
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, 100044, China
| | - Xiaojuan Wang
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, 100044, China
| | - Shuguang Li
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, 100044, China
| | - Yawei Zhang
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, 100044, China
| | - Rhys A Farrer
- UCL Genetics Institute, University College London, Gower Street, London, WC1E 6BT, UK
- Medical Research Council Centre for Medical Mycology at the University of Exeter, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, EX4 4QD, UK
| | - Hui Wang
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, 100044, China.
| | - Francois Balloux
- UCL Genetics Institute, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
36
|
Lin X, He J, Li W, Qi Y, Hu H, Zhang D, Xu F, Chen X, Zhou M. Lung-Targeting Lysostaphin Microspheres for Methicillin-Resistant Staphylococcus aureus Pneumonia Treatment and Prevention. ACS NANO 2021; 15:16625-16641. [PMID: 34582183 DOI: 10.1021/acsnano.1c06460] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Multifunctional antimicrobial strategies are urgently needed to treat methicillin-resistant Staphylococcus aureus (MRSA) caused pneumonia due to its increasing resistance, enhanced virulence, and high pathogenicity. Here, we report that lysostaphin, a bacteriolytic enzyme, encapsulated within poly(lactic-co-glycolic acid) microspheres (LyIR@MS) specially treats planktonic MRSA bacteria, mature biofilms, and related pneumonia. Optimized LyIR@MS with suitable diameters could deliver a sufficient amount of lysostaphin to the lung without a decrease in survival rate after intravenous injection. Furthermore, the degradable properties of the carrier make it safe for targeted release of lysostaphin to eliminate MRSA, repressing the expression of virulence genes and improving the sensitivity of biofilms to host neutrophils. In the MRSA pneumonia mouse model, treatment or prophylaxis with LyIR@MS significantly improved survival rate and relieved inflammatory injury without introducing adverse events. These findings suggest the clinical translational potential of LyIR@MS for the treatment of MRSA-infected lung diseases.
Collapse
Affiliation(s)
- Xiuhui Lin
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Jian He
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310009, China
| | - Wanlin Li
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yuchen Qi
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310009, China
| | - Huiqun Hu
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Dongxiao Zhang
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310009, China
| | - Feng Xu
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Min Zhou
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310009, China
- State Key Laboratory of Modern Optical Instrumentations, Zhejiang University, Hangzhou 310058, China
- Cancer Center, Zhejiang University, Hangzhou 310009, China
| |
Collapse
|
37
|
Aboushanab SA, El-Far AH, Narala VR, Ragab RF, Kovaleva EG. Potential therapeutic interventions of plant-derived isoflavones against acute lung injury. Int Immunopharmacol 2021; 101:108204. [PMID: 34619497 DOI: 10.1016/j.intimp.2021.108204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/19/2021] [Accepted: 09/25/2021] [Indexed: 12/24/2022]
Abstract
Acute lung injury (ALI) is a life-threatening syndrome that possibly leads to high morbidity and mortality as no therapy exists. Several natural ingredients with negligible adverse effects have recently been investigated to possibly inhibit the inflammatory pathways associated with ALI at the molecular level. Isoflavones, as phytoestrogenic compounds, are naturally occurring bioactive compounds that represent the most abundant category of plant polyphenols (Leguminosae family). A broad range of therapeutic activities of isoflavones, including antioxidants, chemopreventive, anti-inflammatory, antiallergic and antibacterial potentials, have been extensively documented in the literature. Our review exclusively focuses on the possible anti-inflammatory, antioxidant role of botanicals'-derived isoflavones against ALI and their immunomodulatory effect in experimentally induced ALI. Despite the limited scope covering their molecular mechanisms, isoflavones substantially contributed to protecting from ALI via inhibiting toll-like receptor 4 (TLR4)/Myd88/NF-κB pathway and subsequent cytokines, chemokines, and adherent proteins. Nonetheless, future research is suggested to fill the gap in elucidating the protective roles of isoflavones to alleviate ALI concerning antioxidant potentials, inhibition of the inflammatory pathways, and associated molecular mechanisms.
Collapse
Affiliation(s)
- Saied A Aboushanab
- Institute of Chemical Engineering, Ural Federal University named after the First President of Russia B. N. Yeltsin, 620002, 19 Mira Yekaterinburg, Russia.
| | - Ali H El-Far
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt; Scientific Chair of Yousef Abdullatif Jameel of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia.
| | | | - Rokia F Ragab
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt.
| | - Elena G Kovaleva
- Institute of Chemical Engineering, Ural Federal University named after the First President of Russia B. N. Yeltsin, 620002, 19 Mira Yekaterinburg, Russia.
| |
Collapse
|
38
|
Liang B, Liang X, Gao F, Long Y, Mai J, Ai X, Wang J, Gao X, Xiong Z, Liang Z, Zhang C, Gong S, Zhou Z. Active Surveillance, Drug Resistance, and Genotypic Profiling of Staphylococcus aureus Among School-Age Children in China. Front Med (Lausanne) 2021; 8:701494. [PMID: 34447764 PMCID: PMC8382981 DOI: 10.3389/fmed.2021.701494] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/09/2021] [Indexed: 12/11/2022] Open
Abstract
Methicillin-susceptible (MSSA) and methicillin-resistant Staphylococcus aureus (MRSA) nasal colonization predisposes individuals for endogenous infections and is a major threat to children. Recently, oxacillin/cefoxitin-susceptible mecA-positive S. aureus (OS-MRSA) has been reported worldwide. Herein, a prospective, cross-sectional study was conducted across five schools, representing three educational stages, in Guangzhou, China. Nasal swabs from 2,375 students were cultured for S. aureus and all isolates were subjected to antibiotic susceptibility testing phenotypically and confirmed by femB and mecA genetic detection; all the isolates were classified as MSSA, MRSA, or OS-MRSA. All strains were also analyzed by multi-locus sequence typing. Among the 2,375 swabs, S. aureus was detected in 744 children (31.3%, 95% CI: 25.9–36.7%), of whom 72 had MRSA (3.0%, 95% CI: 0.6–5.4%) and 4 had OS-MRSA (0.2%, 95% CI: 0.1–0.3%), of which an oxacillin- and cefoxitin-susceptible MRSA strain was identified. The prevalence of S. aureus and MRSA was higher in younger children. The highest percentage of drug resistance of the S. aureus isolates (n = 744) was to penicillin (85.5%), followed by erythromycin (43.3%) and clidamycin (41.0%). The most prevalent sequence types (STs) were ST30, ST45, and ST188 in MSSA, accounting for 38.7% of the total isolates, whereas ST45, ST59, and ST338 accounted for 74.6% of the MRSA isolates and ST338 accounted for 50.0% of the OS-MRSA isolates. The MRSA and OS-MRSA isolates (n = 76) were grouped into three clades and one singleton, with clonal complex (CC) 45 as the most predominant linkage. The top nine multi-locus sequence typing-based CCs (CC30, CC45, CC5, CC1, CC15, CC944, CC398, CC59, CC7) represented 86.7% of all S. aureus isolates. All CC30 isolates were resistant to erythromycin and clidamycin, and almost all these isolates were also resistant to penicillin (99.2%). The CC45 and CC59 isolates exhibited high resistance rates to oxacillin at 31.5 and 59.0%, respectively. This study provides updated data valuable for designing effective control strategies to mitigate the burden of disease and to improve the adequacy of empirical antimicrobial treatments for potentially harmful infections.
Collapse
Affiliation(s)
- Bingshao Liang
- Clinical Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiaoyun Liang
- Clinical Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Fei Gao
- Clinical Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yan Long
- Clinical Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jialiang Mai
- Clinical Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiaolan Ai
- Clinical Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jielin Wang
- Clinical Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiurong Gao
- Clinical Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Zhile Xiong
- Clinical Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Zhuwei Liang
- Clinical Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Chao Zhang
- Clinical Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Sitang Gong
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Zhenwen Zhou
- Clinical Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,Department of Laboratory Medicine, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
39
|
Zhang W, Ren J, Lu J, Li P, Zhang W, Wang H, Tang B. Elucidating the Relationship between ROS and Protein Phosphorylation through In Situ Fluorescence Imaging in the Pneumonia Mice. Anal Chem 2021; 93:10907-10915. [PMID: 34324298 DOI: 10.1021/acs.analchem.1c01690] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Revealing the relationship between reactive oxygen species (ROS) and levels of protein phosphorylation is of great significance for understanding the pathogenesis of diseases. Although mass spectrometry is used as a classical method for protein phosphorylation analysis, there are still some challenges to realize in vivo protein phosphorylation recognition. Herein, we designed and prepared an metal-organic framework (MOF)-based fluorescent nanoprobe with Zr(IV) and boronate ester as an active center, which achieved simultaneous recognition of ROS and phosphorylation sites. The ROS unit was constructed by 1,8-naphthalimide and boronate ester as a fluorophore and a recognition group, respectively. The specific interaction between Zr(IV) and a phosphate group was used to realize fluorescence imaging of phosphorylation sites. Using the advantages of two-photon property of the ROS recognition unit, the nanoprobe can effectively reduce the background fluorescence and thus improve the imaging sensitivity. Finally, the MOF-based nanoprobe was successfully applied to reveal the relationship between ROS and levels of phosphorylation in pneumonia mice, which illustrated that the ROS and phosphorylation levels in the process of pulmonary inflammation were obviously higher than those of the normal mice. This work provides feasible fluorescence tools that have important significance for revealing pathogenesis of diseases.
Collapse
Affiliation(s)
- Wei Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, P. R. China
| | - Jie Ren
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, P. R. China
| | - Jun Lu
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, P. R. China
| | - Ping Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, P. R. China
| | - Wen Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, P. R. China
| | - Hui Wang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, P. R. China
| |
Collapse
|
40
|
A Fragile Balance: Does Neutrophil Extracellular Trap Formation Drive Pulmonary Disease Progression? Cells 2021; 10:cells10081932. [PMID: 34440701 PMCID: PMC8394734 DOI: 10.3390/cells10081932] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/22/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
Neutrophils act as the first line of defense during infection and inflammation. Once activated, they are able to fulfil numerous tasks to fight inflammatory insults while keeping a balanced immune response. Besides well-known functions, such as phagocytosis and degranulation, neutrophils are also able to release "neutrophil extracellular traps" (NETs). In response to most stimuli, the neutrophils release decondensed chromatin in a NADPH oxidase-dependent manner decorated with histones and granule proteins, such as neutrophil elastase, myeloperoxidase, and cathelicidins. Although primarily supposed to prevent microbial dissemination and fight infections, there is increasing evidence that an overwhelming NET response correlates with poor outcome in many diseases. Lung-related diseases especially, such as bacterial pneumonia, cystic fibrosis, chronic obstructive pulmonary disease, aspergillosis, influenza, and COVID-19, are often affected by massive NET formation. Highly vascularized areas as in the lung are susceptible to immunothrombotic events promoted by chromatin fibers. Keeping this fragile equilibrium seems to be the key for an appropriate immune response. Therapies targeting dysregulated NET formation might positively influence many disease progressions. This review highlights recent findings on the pathophysiological influence of NET formation in different bacterial, viral, and non-infectious lung diseases and summarizes medical treatment strategies.
Collapse
|
41
|
Zeouk I, Ouedrhiri W, Sifaoui I, Bazzocchi IL, Piñero JE, Jiménez IA, Lorenzo-Morales J, Bekhti K. Bioguided Isolation of Active Compounds from Rhamnus alaternus against Methicillin-Resistant Staphylococcus aureus (MRSA) and Panton-Valentine Leucocidin Positive Strains (MSSA-PVL). Molecules 2021; 26:molecules26144352. [PMID: 34299627 PMCID: PMC8306708 DOI: 10.3390/molecules26144352] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 01/24/2023] Open
Abstract
Despite intensified efforts to develop an effective antibiotic, S. aureus is still a major cause of mortality and morbidity worldwide. The multidrug resistance of bacteria has considerably increased the difficulties of scientific research and the concomitant emergence of resistance is to be expected. In this study we have investigated the in vitro activity of 15 ethanol extracts prepared from Moroccan medicinal plants traditionally used for treatment of skin infections. Among the tested species I. viscosa, C. oxyacantha, R. tinctorum, A. herba alba, and B. hispanica showed moderate anti-staphylococcal activity. However, R. alaternus showed promising growth-inhibitory effects against specific pathogenic bacteria especially methicillin-susceptible Staphylococcus aureus Panton-Valentine leucocidin positive (MSSA-PVL) and methicillin-resistant S. aureus (MRSA). The bioguided fractionation of this plant using successive chromatographic separations followed by nuclear magnetic resonance (NMR) and mass spectrometry (MS) including EIMS and HREIMS analysis yielded the emodin (1) and kaempferol (2). Emodin being the most active with MICs ranging between 15.62 and 1.95 µg/mL and showing higher activity against the tested strains in comparison with the crude extract, its mechanism of action and the structure-activity relationship were interestingly discussed. The active compound has not displayed toxicity toward murine macrophage cells. The results obtained in the current study support the traditional uses of R. alaternus and suggest that this species could be a good source for the development of new anti-staphylococcal agents.
Collapse
Affiliation(s)
- Ikrame Zeouk
- Instituto Universitario De Enfermedades Tropicales y Salud Pública de Canarias, Universidad de la Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203 La Laguna, Spain;
- Laboratory of Microbial Biotechnology and Bioactive Molecules, Department of Biology, Faculty of Sciences and Techniques, Sidi Mohamed Ben Abdellah University, Fez 2202, Morocco;
- Correspondence: (I.Z.); (J.E.P.); (J.L.-M.); Tel.: +212-621-290-377 (I.Z.); +349-22-316-502 (J.E.P.); +349-22-318-402 (J.L.-M.)
| | - Wessal Ouedrhiri
- Laboratory of Engineering, Electrochemistry, Modeling and Environment, Department of Chemistry, Faculty of Science, Sidi Mohamed Ben Abdellah University, Fez 2202, Morocco;
| | - Ines Sifaoui
- Instituto Universitario De Enfermedades Tropicales y Salud Pública de Canarias, Universidad de la Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203 La Laguna, Spain;
- Departamento de Obstetricia, Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de la Laguna, 38203 La Laguna, Spain
- Red de Investigación Colaborativa en Enfermedades Tropicales (RICET), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Isabel L. Bazzocchi
- Departamento de Química Orgánica, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez 2, 38206 La Laguna, Spain; (I.L.B.); (I.A.J.)
| | - José E. Piñero
- Instituto Universitario De Enfermedades Tropicales y Salud Pública de Canarias, Universidad de la Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203 La Laguna, Spain;
- Departamento de Obstetricia, Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de la Laguna, 38203 La Laguna, Spain
- Red de Investigación Colaborativa en Enfermedades Tropicales (RICET), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (I.Z.); (J.E.P.); (J.L.-M.); Tel.: +212-621-290-377 (I.Z.); +349-22-316-502 (J.E.P.); +349-22-318-402 (J.L.-M.)
| | - Ignacio A. Jiménez
- Departamento de Química Orgánica, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, Avenida Astrofísico Francisco Sánchez 2, 38206 La Laguna, Spain; (I.L.B.); (I.A.J.)
| | - Jacob Lorenzo-Morales
- Instituto Universitario De Enfermedades Tropicales y Salud Pública de Canarias, Universidad de la Laguna, Avda. Astrofísico Fco. Sánchez, S/N, 38203 La Laguna, Spain;
- Departamento de Obstetricia, Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de la Laguna, 38203 La Laguna, Spain
- Red de Investigación Colaborativa en Enfermedades Tropicales (RICET), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (I.Z.); (J.E.P.); (J.L.-M.); Tel.: +212-621-290-377 (I.Z.); +349-22-316-502 (J.E.P.); +349-22-318-402 (J.L.-M.)
| | - Khadija Bekhti
- Laboratory of Microbial Biotechnology and Bioactive Molecules, Department of Biology, Faculty of Sciences and Techniques, Sidi Mohamed Ben Abdellah University, Fez 2202, Morocco;
| |
Collapse
|
42
|
An AI-based auxiliary empirical antibiotic therapy model for children with bacterial pneumonia using low-dose chest CT images. Jpn J Radiol 2021; 39:973-983. [PMID: 34101118 PMCID: PMC8490241 DOI: 10.1007/s11604-021-01136-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/13/2021] [Indexed: 11/27/2022]
Abstract
Purpose To construct an auxiliary empirical antibiotic therapy (EAT) multi-class classification model for children with bacterial pneumonia using radiomics features based on artificial intelligence and low-dose chest CT images. Materials and methods Data were retrospectively collected from children with pathogen-confirmed bacterial pneumonia including Gram-positive bacterial pneumonia (122/389, 31%), Gram-negative bacterial pneumonia (159/389, 41%) and atypical bacterial pneumonia (108/389, 28%) from January 1 to June 30, 2019. Nine machine-learning models were separately evaluated based on radiomics features extracted from CT images; three optimal submodels were constructed and integrated to form a multi-class classification model. Results We selected five features to develop three radiomics submodels: a Gram-positive model, a Gram-negative model and an atypical model. The comprehensive radiomics model using support vector machine method yielded an average area under the curve (AUC) of 0.75 [95% confidence interval (CI), 0.65–0.83] and accuracy (ACC) of 0.58 [sensitivity (SEN), 0.57; specificity (SPE), 0.78] in the training set, and an average AUC of 0.73 (95% CI 0.61–0.79) and ACC of 0.54 (SEN, 0.52; SPE, 0.75) in the test set. Conclusion This auxiliary EAT radiomics multi-class classification model was deserved to be researched in differential diagnosing bacterial pneumonias in children.
Collapse
|
43
|
Abstract
Staphylococcus aureus is both a commensal and a pathogenic bacterium for humans. Its ability to induce severe infections is based on a wide range of virulence factors. S. aureus community-acquired pneumonia (SA-CAP) is rare and severe, and the contribution of certain virulence factors in this disease has been recognized over the past 2 decades. First, the factors involved in metabolism adaptation are crucial for S. aureus survival in the lower respiratory tract, and toxins and enzymes are required for it to cross the pulmonary epithelial barrier. S. aureus subsequently faces host defense mechanisms, including the epithelial barrier, but most importantly the immune system. Here, again, S. aureus uses myriad virulence factors to successfully escape from the host's defenses and takes advantage of them. The impact of S. aureus virulence, combined with the collateral damage caused by an overwhelming immune response, leads to severe tissue damage and adverse clinical outcomes. In this review, we summarize step by step all of the S. aureus factors implicated in CAP and described to date, and we provide an outlook for future research.
Collapse
Affiliation(s)
- Mariane Pivard
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Karen Moreau
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - François Vandenesch
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- Centre National de Référence des Staphylocoques, Institut des agents infectieux, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
44
|
Mraheil MA, Toque HA, La Pietra L, Hamacher J, Phanthok T, Verin A, Gonzales J, Su Y, Fulton D, Eaton DC, Chakraborty T, Lucas R. Dual Role of Hydrogen Peroxide as an Oxidant in Pneumococcal Pneumonia. Antioxid Redox Signal 2021; 34:962-978. [PMID: 32283950 PMCID: PMC8035917 DOI: 10.1089/ars.2019.7964] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Significance:Streptococcus pneumoniae (Spn), a facultative anaerobic Gram-positive human pathogen with increasing rates of penicillin and macrolide resistance, is a major cause of lower respiratory tract infections worldwide. Pneumococci are a primary agent of severe pneumonia in children younger than 5 years and of community-acquired pneumonia in adults. A major defense mechanism toward Spn is the generation of reactive oxygen species, including hydrogen peroxide (H2O2), during the oxidative burst of neutrophils and macrophages. Paradoxically, Spn produces high endogenous levels of H2O2 as a strategy to promote colonization. Recent Advances: Pneumococci, which express neither catalase nor common regulators of peroxide stress resistance, have developed unique mechanisms to protect themselves from H2O2. Spn generates high levels of H2O2 as a strategy to promote colonization. Production of H2O2 moreover constitutes an important virulence phenotype and its cellular activities overlap and complement those of other virulence factors, such as pneumolysin, in modulating host immune responses and promoting organ injury. Critical Issues: This review examines the dual role of H2O2 in pneumococcal pneumonia, from the viewpoint of both the pathogen (defense mechanisms, lytic activity toward competing pathogens, and virulence) and the resulting host-response (inflammasome activation, endoplasmic reticulum stress, and damage to the alveolar-capillary barrier in the lungs). Future Directions: An understanding of the complexity of H2O2-mediated host-pathogen interactions is necessary to develop novel strategies that target these processes to enhance lung function during severe pneumonia.
Collapse
Affiliation(s)
- Mobarak Abu Mraheil
- Institute for Medical Microbiology, Justus-Liebig University, Giessen, Germany
| | - Haroldo A Toque
- Vascular Biology Center and Medical College of Georgia at Augusta University, Augusta, Georgia, USA.,Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Luigi La Pietra
- Institute for Medical Microbiology, Justus-Liebig University, Giessen, Germany
| | - Juerg Hamacher
- Internal Medicine and Pneumology, Lindenhofspital, Bern, Switzerland.,Lungen- und Atmungsstiftung Bern, Bern, Switzerland.,Internal Medicine V-Pneumology, Allergology, Respiratory and Environmental Medicine, Faculty of Medicine, Saarland University, Saarbrücken, Germany
| | - Tenzing Phanthok
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Alexander Verin
- Vascular Biology Center and Medical College of Georgia at Augusta University, Augusta, Georgia, USA.,Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Joyce Gonzales
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Yunchao Su
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - David Fulton
- Vascular Biology Center and Medical College of Georgia at Augusta University, Augusta, Georgia, USA.,Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Douglas C Eaton
- Department of Medicine, Emory School of Medicine, Atlanta, Georgia, USA
| | - Trinad Chakraborty
- Institute for Medical Microbiology, Justus-Liebig University, Giessen, Germany
| | - Rudolf Lucas
- Vascular Biology Center and Medical College of Georgia at Augusta University, Augusta, Georgia, USA.,Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, Georgia, USA.,Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| |
Collapse
|
45
|
Abstract
Pneumonia is a common acute respiratory infection that affects the alveoli and distal airways; it is a major health problem and associated with high morbidity and short-term and long-term mortality in all age groups worldwide. Pneumonia is broadly divided into community-acquired pneumonia or hospital-acquired pneumonia. A large variety of microorganisms can cause pneumonia, including bacteria, respiratory viruses and fungi, and there are great geographical variations in their prevalence. Pneumonia occurs more commonly in susceptible individuals, including children of <5 years of age and older adults with prior chronic conditions. Development of the disease largely depends on the host immune response, with pathogen characteristics having a less prominent role. Individuals with pneumonia often present with respiratory and systemic symptoms, and diagnosis is based on both clinical presentation and radiological findings. It is crucial to identify the causative pathogens, as delayed and inadequate antimicrobial therapy can lead to poor outcomes. New antibiotic and non-antibiotic therapies, in addition to rapid and accurate diagnostic tests that can detect pathogens and antibiotic resistance will improve the management of pneumonia.
Collapse
|
46
|
Hemoglobin Induces Early and Robust Biofilm Development in Streptococcus pneumoniae by a Pathway That Involves comC but Not the Cognate comDE Two-Component System. Infect Immun 2021; 89:IAI.00779-20. [PMID: 33397818 DOI: 10.1128/iai.00779-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 11/20/2022] Open
Abstract
Streptococcus pneumoniae grows in biofilms during both asymptomatic colonization and infection. Pneumococcal biofilms on abiotic surfaces exhibit delayed growth and lower biomass and lack the structures seen on epithelial cells or during nasopharyngeal carriage. We show here that adding hemoglobin to the medium activated unusually early and vigorous biofilm growth in multiple S. pneumoniae serotypes grown in batch cultures on abiotic surfaces. Human blood (but not serum, heme, or iron) also stimulated biofilms, and the pore-forming pneumolysin, ply, was required for this induction. S. pneumoniae transitioning from planktonic into sessile growth in the presence of hemoglobin displayed an extensive transcriptome remodeling within 1 and 2 h. Differentially expressed genes included those involved in the metabolism of carbohydrates, nucleotides, amino acid, and lipids. The switch into adherent states also influenced the expression of several regulatory systems, including the comCDE genes. Inactivation of comC resulted in 67% reduction in biofilm formation, while the deletion of comD or comE had limited or no effect, respectively. These observations suggest a novel route for CSP-1 signaling independent of the cognate ComDE two-component system. Biofilm induction and the associated transcriptome remodeling suggest hemoglobin serves as a signal for host colonization in pneumococcus.
Collapse
|
47
|
Barman TK, Racine R, Bonin JL, Califano D, Salmon SL, Metzger DW. Sequential targeting of interferon pathways for increased host resistance to bacterial superinfection during influenza. PLoS Pathog 2021; 17:e1009405. [PMID: 33690728 PMCID: PMC7978370 DOI: 10.1371/journal.ppat.1009405] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 03/19/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022] Open
Abstract
Bacterial co-infections represent a major clinical complication of influenza. Host-derived interferon (IFN) increases susceptibility to bacterial infections following influenza, but the relative roles of type-I versus type-II IFN remain poorly understood. We have used novel mouse models of co-infection in which colonizing pneumococci were inoculated into the upper respiratory tract; subsequent sublethal influenza virus infection caused the bacteria to enter the lungs and mediate lethal disease. Compared to wild-type mice or mice deficient in only one pathway, mice lacking both IFN pathways demonstrated the least amount of lung tissue damage and mortality following pneumococcal-influenza virus superinfection. Therapeutic neutralization of both type-I and type-II IFN pathways similarly provided optimal protection to co-infected wild-type mice. The most effective treatment regimen was staggered neutralization of the type-I IFN pathway early during co-infection combined with later neutralization of type-II IFN, which was consistent with the expression and reported activities of these IFNs during superinfection. These results are the first to directly compare the activities of type-I and type-II IFN during superinfection and provide new insights into potential host-directed targets for treatment of secondary bacterial infections during influenza. Bacterial co-infections represent a common and challenging clinical complication of influenza. Type-I and type-II interferon (IFN) pathways enhance susceptibility to influenza-pneumococcal co-infection, leading to increased lung pathology and mortality. However, the comparative importance of type-I versus type-II IFN remains unclear. We have used two novel mouse models of co-infection in which pneumococci were inoculated into the upper respiratory tract followed two days later by influenza virus infection. Virus co-infection caused IFN-dependent inflammation that facilitated spreading of the colonizing bacteria into the lungs, followed by tissue damage and death. In this pneumococcal-influenza virus superinfection model, mice lacking both type-I and type-II IFN pathways demonstrated minimal lung pathology and increased survival compared to wild-type mice and mice deficient in only one pathway. Therapeutic neutralization of both type-I and type-II IFN pathways similarly provided optimal protection to superinfected wild-type mice. The most effective treatment regimen involved neutralization of the type-I IFN pathway early during co-infection combined with later neutralization of the type-II IFN pathway. These results provide new insights into potential host-directed therapy for management of bacterial-viral superinfections.
Collapse
Affiliation(s)
- Tarani Kanta Barman
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Rachael Racine
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Jesse L. Bonin
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Danielle Califano
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Sharon L. Salmon
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Dennis W. Metzger
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
- * E-mail:
| |
Collapse
|
48
|
Zeng S, Chen D, Liu G, Wu YX, Gao ZQ, Su Y, Yuan JN, Liu L, Shan JC, Pang QF, Zhu T. Salvinorin A protects against methicillin resistant staphylococcus aureus-induced acute lung injury via Nrf2 pathway. Int Immunopharmacol 2020; 90:107221. [PMID: 33293260 DOI: 10.1016/j.intimp.2020.107221] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 02/05/2023]
Abstract
Salvinorin A (SA), a neoclerodane diterpene, is isolated from the dried leaves ofSalvia divinorum. SA has traditionally been used treatments for chronic pain diseases. Recent research has demonstrated that SA possesses the anti-inflammatory property. The present study aim to explore the effects and potentialmechanisms ofSA in protection against Methicillin Resistant Staphylococcus aureus (MRSA)-induced acute lung injury (ALI). Here, we firstly found that verylowdosesof SA (50 μg/kg) could markedly decrease the infiltration of pulmonary neutrophils, mRNA expression of pro-inflammatory cytokines (TNF-α, IL-1β and IL-6) and then attenuated ALI cause by MRSA infection in mice. In vitro findings revealed that SA attenuated lipoteichoicacid-induced apoptosis, inflammation and oxidative stress in RAW264.7 cells. Mechanism research revealed that SA increased both mRNA levels and protein levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and up-regulated mRNA expression of its downstream genes (HO-1, Gclm, Trx-1, SOD1 and SOD2). Additionally, Nrf2 knockout mice abolished the inhibitory effect of SA on neutrophil accumulation and oxidative stress in MRSA-induced ALI. In conclusion, SA attenuates MRSA-induced ALI via Nrf2 signaling pathways.
Collapse
Affiliation(s)
- Si Zeng
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Anesthesiology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Dan Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Gang Liu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ya-Xian Wu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Zhi-Qi Gao
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ying Su
- Library, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jia-Ning Yuan
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Liu Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Anesthesiology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Jia-Chen Shan
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qing-Feng Pang
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China.
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
49
|
Akhter F, Womack E, Vidal JE, Le Breton Y, McIver KS, Pawar S, Eichenbaum Z. Hemoglobin stimulates vigorous growth of Streptococcus pneumoniae and shapes the pathogen's global transcriptome. Sci Rep 2020; 10:15202. [PMID: 32938947 PMCID: PMC7494912 DOI: 10.1038/s41598-020-71910-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/20/2020] [Indexed: 12/14/2022] Open
Abstract
Streptococcus pneumoniae (Spn) must acquire iron from the host to establish infection. We examined the impact of hemoglobin, the largest iron reservoir in the body, on pneumococcal physiology. Supplementation with hemoglobin allowed Spn to resume growth in an iron-deplete medium. Pneumococcal growth with hemoglobin was unusually robust, exhibiting a prolonged logarithmic growth, higher biomass, and extended viability in both iron-deplete and standard medium. We observed the hemoglobin-dependent response in multiple serotypes, but not with other host proteins, free iron, or heme. Remarkably, hemoglobin induced a sizable transcriptome remodeling, effecting virulence and metabolism in particular genes facilitating host glycoconjugates use. Accordingly, Spn was more adapted to grow on the human α − 1 acid glycoprotein as a sugar source with hemoglobin. A mutant in the hemoglobin/heme-binding protein Spbhp-37 was impaired for growth on heme and hemoglobin iron. The mutant exhibited reduced growth and iron content when grown in THYB and hemoglobin. In summary, the data show that hemoglobin is highly beneficial for Spn cultivation in vitro and suggest that hemoglobin might drive the pathogen adaptation in vivo. The hemoglobin receptor, Spbhp-37, plays a role in mediating the positive influence of hemoglobin. These novel findings provide intriguing insights into pneumococcal interactions with its obligate human host.
Collapse
Affiliation(s)
- Fahmina Akhter
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Edroyal Womack
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Jorge E Vidal
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yoann Le Breton
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park (UMCP), College Park, MD, USA.,Wound Infections Department, Bacterial Diseases Branch, The Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Kevin S McIver
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park (UMCP), College Park, MD, USA
| | - Shrikant Pawar
- Department of Biology, Georgia State University, Atlanta, GA, USA.,Yale Center for Genome Analysis, Yale University, New Haven, CT, USA
| | | |
Collapse
|
50
|
Influenza sequelae: from immune modulation to persistent alveolitis. Clin Sci (Lond) 2020; 134:1697-1714. [PMID: 32648583 DOI: 10.1042/cs20200050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/25/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023]
Abstract
Acute influenza virus infections are a global public health concern accounting for millions of illnesses worldwide ranging from mild to severe with, at time, severe complications. Once an individual is infected, the immune system is triggered in response to the pathogen. This immune response can be beneficial ultimately leading to the clearance of the viral infection and establishment of immune memory mechanisms. However, it can be detrimental by increasing susceptibility to secondary bacterial infections and resulting in permanent changes to the lung architecture, in the form of fibrotic sequelae. Here, we review influenza associated bacterial super-infection, the formation of T-cell memory, and persistent lung injury resulting from influenza infection.
Collapse
|