1
|
Ghosh S, Roy S, Baid N, Das UK, Rakshit S, Sanghavi P, Hajra D, Das S, Menon S, Sahil M, Shaw S, Rajmani RS, Adicherla H, Kaledhonkar S, Mondal J, Chakravortty D, Mallik R, Banerjee A. Host AAA-ATPase VCP/p97 lyses ubiquitinated intracellular bacteria as an innate antimicrobial defence. Nat Microbiol 2025; 10:1099-1114. [PMID: 40217128 DOI: 10.1038/s41564-025-01984-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 03/14/2025] [Indexed: 04/16/2025]
Abstract
Cell-autonomous immunity prevents intracellular pathogen growth through mechanisms such as ubiquitination and proteasomal targeting of bacteria for degradation. However, how the proteasome eradicates ubiquitinated bacteria has remained unclear. Here we show that host AAA-ATPase, VCP/p97, associates with diverse cytosol-exposed ubiquitinated bacteria (Streptococcus pneumoniae, Salmonella enterica serovar Typhimurium, Streptococcus pyogenes) and requires the ATPase activity in its D2 domain to reduce intracellular bacterial loads. Combining optical trap approaches along with molecular dynamic simulations, in vitro reconstitution and immunogold transmission electron microscopy, we demonstrate that p97 applies mechanical force to extract ubiquitinated surface proteins, BgaA and PspA, from S. pneumoniae cell membranes. This causes extensive membrane lysis and release of cytosolic content, thereby killing the pathogen. Further, p97 also controls S. pneumoniae proliferation in mice, ultimately protecting from fatal sepsis. Overall, we discovered a distinct innate antimicrobial function of p97 that can protect the host against lethal bacterial infections.
Collapse
Affiliation(s)
- Sourav Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
| | - Suvapriya Roy
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
| | - Navin Baid
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
| | - Udit Kumar Das
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
| | - Sumit Rakshit
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
| | - Paulomi Sanghavi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
| | - Dipasree Hajra
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Sayani Das
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
| | - Sneha Menon
- Tata Institute of Fundamental Research, Hyderabad, Telangana, India
| | - Mohammad Sahil
- Tata Institute of Fundamental Research, Hyderabad, Telangana, India
| | - Sudipti Shaw
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
| | - Raju S Rajmani
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Harikrishna Adicherla
- CSIR-Centre for Cellular and Molecular Biology, Habsiguda, Hyderabad, Telangana, India
| | - Sandip Kaledhonkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
| | - Jagannath Mondal
- Tata Institute of Fundamental Research, Hyderabad, Telangana, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Roop Mallik
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India
| | - Anirban Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India.
| |
Collapse
|
2
|
Mazzuoli MV, van Raaphorst R, Martin L, Bock F, Thierry A, Marbouty M, Waclawiková B, Stinenbosch J, Koszul R, Veening JW. HU promotes higher order chromosome organization and influences DNA replication rates in Streptococcus pneumoniae. Nucleic Acids Res 2025; 53:gkaf312. [PMID: 40263708 PMCID: PMC12014288 DOI: 10.1093/nar/gkaf312] [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: 10/04/2024] [Revised: 03/17/2025] [Accepted: 04/09/2025] [Indexed: 04/24/2025] Open
Abstract
Nucleoid-associated proteins (NAPs) are crucial for maintaining chromosomal compaction and architecture, and are actively involved in DNA replication, recombination, repair, and gene regulation. In Streptococcus pneumoniae, the role of the highly conserved NAP HU in chromosome conformation has not yet been investigated. Here, we use a multi-scale approach to explore HU's role in chromosome conformation and segregation dynamics. By combining superresolution microscopy and whole-genome-binding analysis, we describe the nucleoid as a dynamic structure where HU binds transiently across the entire nucleoid, with a preference for the origin of replication over the terminus. Reducing cellular HU levels impacts nucleoid maintenance and disrupts nucleoid scaling with cell size, similar to the distortion caused by fluoroquinolones, supporting its requirement for maintaining DNA supercoiling. Furthermore, in cells lacking HU, the replication machinery is misplaced, preventing cells from initiating and proceeding with ongoing replication. Chromosome conformation capture coupled to deep sequencing (Hi-C) revealed that HU is required to maintain cohesion between the two chromosomal arms, similar to the structural maintenance of chromosome complex. Together, we show that by promoting long-range chromosome interactions and supporting the architecture of the domain encompassing the origin, HU is essential for chromosome integrity and the intimately related processes of replication and segregation.
Collapse
Affiliation(s)
- Maria-Vittoria Mazzuoli
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, Lausanne, CH-1015, Switzerland
| | - Renske van Raaphorst
- Department of Molecular Microbiology, Groningen Institute of Biomolecular Sciences & Biotechnology, University of Groningen, 9747, The Netherlands
| | - Louise S Martin
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, Lausanne, CH-1015, Switzerland
| | - Florian P Bock
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, Lausanne, CH-1015, Switzerland
| | - Agnès Thierry
- Institut Pasteur, CNRS UMR3525, Université Paris Cité, Unité Régulation Spatiale des Génomes, Paris, 75015, France
| | - Martial Marbouty
- Institut Pasteur, CNRS UMR3525, Université Paris Cité, Unité Régulation Spatiale des Génomes, Paris, 75015, France
| | - Barbora Waclawiková
- Department of Molecular Microbiology, Groningen Institute of Biomolecular Sciences & Biotechnology, University of Groningen, 9747, The Netherlands
| | - Jasper Stinenbosch
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, Lausanne, CH-1015, Switzerland
| | - Romain Koszul
- Institut Pasteur, CNRS UMR3525, Université Paris Cité, Unité Régulation Spatiale des Génomes, Paris, 75015, France
| | - Jan-Willem Veening
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Biophore Building, Lausanne, CH-1015, Switzerland
| |
Collapse
|
3
|
Tian Y, Jiang X, Bao C, Abdelaal T, Chen D, Wang W, Li F, Lei L, Li N. Mass cytometry analysis reveals a cross-tissue immune landscape in Actinobacillus pleuropneumoniae-induced pneumonia. Microbiol Spectr 2025:e0266524. [PMID: 40237529 DOI: 10.1128/spectrum.02665-24] [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: 10/23/2024] [Accepted: 03/13/2025] [Indexed: 04/18/2025] Open
Abstract
Porcine contagious pleuropneumonia caused by Actinobacillus pleuropneumoniae (APP) is a fatal respiratory disease that threatens the worldwide farming industry's health. The immune responses of extrapulmonary tissues play an important role in developing porcine contagious pleuropneumonia; however, the immune responses of extrapulmonary tissues induced by APP are rarely uncovered. Here, we used high-dimensional mass cytometry to investigate the immune cell response in the spleen and peripheral blood during APP infection in mice. We found that the immune response triggered by APP was highly tissue-specific. Numerous infection time- or tissue-specific immune cell clusters, including previously unrecognized ones, were also identified in the spleen and peripheral blood. Integrative analysis of splenic lymphoid and myeloid cell clusters maps the dynamic immune response cellular network during APP infection. Surprisingly, during the early stages of APP infection, the majority of the top 6 cell clusters contributing to the infection time-specificity in the spleen were adaptive immune cell clusters rather than innate immune cell clusters, among which CD24hiMHCII+CD8+TEM cells exhibited a stronger expression of IFN-γ, IL-17A, and IL-10 compared to the CD24lo compartment. In peripheral blood, there was unprecedented heterogeneity in the immune cell composition. Also, peripheral immune cell clusters closely related to the severity of APP infection were identified. In summary, our data provide a systemic and comprehensive overview of the immune responses to APP infection in the spleen and peripheral blood. This provides a foundation for understanding the immune pathogenesis of APP and identifying potential diagnostic biomarkers and therapeutic targets. IMPORTANCE This study explored the cross-tissue immune dynamic landscape in the APP-induced pneumonia model by utilizing high-dimensional mass cytometry. We discovered that APP-induced immune responses are tissue-specific. Key infection-specific clusters in the spleen and peripheral blood were identified, some of which were previously unrecognized. Meanwhile, the specific functions of APP infection-related immune subsets were explored. The research systematically outlined an overview of immune responses in these tissues, deepening the understanding of APP pathogenesis and laying the foundation for the search for diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Yanyan Tian
- Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xuan Jiang
- Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Chuntong Bao
- Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Tamin Abdelaal
- Leiden Computational Biology Center, Leiden University Medical Center, Leiden, Netherlands
- Department of Pattern Recognition and Bioinformatics Group, Delft University of Technology, Delft, Netherlands
| | - Dexi Chen
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, Beijing, China
| | - Wenjing Wang
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, Beijing, China
| | - Fengyang Li
- Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Liancheng Lei
- Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Na Li
- Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
4
|
García E. Structure, Function, and Regulation of LytA: The N-Acetylmuramoyl-l-alanine Amidase Driving the "Suicidal Tendencies" of Streptococcus pneumoniae-A Review. Microorganisms 2025; 13:827. [PMID: 40284663 PMCID: PMC12029793 DOI: 10.3390/microorganisms13040827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
Streptococcus pneumoniae (pneumococcus) is a significant human pathogen responsible for a range of diseases from mild infections to invasive pneumococcal diseases, particularly affecting children, the elderly, and immunocompromised individuals. Despite pneumococcal conjugate vaccines having reduced disease incidence, challenges persist due to serotype diversity, vaccine coverage gaps, and antibiotic resistance. This review highlights the role of LytA, a key autolysin (N-acetylmuramoyl-l-alanine amidase), in pneumococcal biology. LytA regulates autolysis, contributes to inflammation, and biofilm formation, and impairs bacterial clearance. It also modulates complement activation, aiding immune evasion. LytA expression is influenced by environmental signals and genetic regulation and is tied to competence for genetic transformation, which is an important virulence trait, particularly in meningitis. With the increase in antibiotic resistance, LytA has emerged as a potential therapeutic target. Current research explores its use in bacteriolytic therapies, vaccine development, and synergistic antibiotic strategies. Various compounds, including synthetic peptides, plant extracts, and small molecules, have been investigated for their ability to trigger LytA-mediated bacterial lysis. Future directions include the development of novel anti-pneumococcal interventions leveraging LytA's properties while overcoming vaccine efficacy and resistance-related challenges. Human challenge models and animal studies continue to deepen our understanding of pneumococcal pathogenesis and potential treatment strategies.
Collapse
Affiliation(s)
- Ernesto García
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, 28040 Madrid, Spain
| |
Collapse
|
5
|
Nguyen TD, Le HD, Dang GC, Jung HS, Choi Y, Khim K, Kim Y, Lee SE, Rhee JH. A combined adjuvant and ferritin nanocage based mucosal vaccine against Streptococcus pneumoniae induces protective immune responses in a murine model. Nat Commun 2025; 16:2871. [PMID: 40128220 PMCID: PMC11933286 DOI: 10.1038/s41467-025-58115-8] [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: 06/04/2024] [Accepted: 03/13/2025] [Indexed: 03/26/2025] Open
Abstract
Protein nanocages are multimeric structures that can be engineered to mimic the molecular conformation of microorganisms. Based on previous findings showing that a mucosal FlaB-tPspA fusion (flagellin fused with truncated PspA antigen of Streptococcus pneumoniae) vaccine-induced protective immune response against S. pneumoniae, we develop a ferritin nanocage vaccine displaying multivalent presentation of both antigen and adjuvant on a nanocarrier using the SpyTag/SpyCatcher strategy. The 1:1 antigen/adjuvant nanocage is further used as a mucosal vaccine, which can translocate to draining lymph nodes with higher efficiency than fusion vaccine. Moreover, intranasal immunization with the nanocage vaccine significantly enhances mucosal immune responses with more efficient B-cell memory generation and antibody maturation, as well as more balanced (Th1/Th2) immune responses with increased IFN-γ and IL-17 production, comparing with fusion vaccine. Mice immunized with the nanocage vaccine exhibited enhanced protection against lethal infection compare to the FlaB-tPspA fusion group. Our study thus demonstrates the effectiveness of an all-in-one nanocage mucosal vaccine platform, which guarantees enhanced protection with balanced immune responses.
Collapse
Affiliation(s)
- Tien Duc Nguyen
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - Hoang Duy Le
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - Giang Chau Dang
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - Hyun Seok Jung
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - Yoonjoo Choi
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - Koemchhoy Khim
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea
| | - Young Kim
- Department of Oral Pathology, Chonnam National University School of Dentistry, Gwangju, Republic of Korea
| | - Shee Eun Lee
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea.
- National Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, Republic of Korea.
- Department of Pharmacology and Dental Therapeutics, Chonnam National University School of Dentistry, Gwangju, Republic of Korea.
| | - Joon Haeng Rhee
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea.
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea.
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, Republic of Korea.
| |
Collapse
|
6
|
Bouma RG, Wang AZ, den Haan JMM. Exploring CD169 + Macrophages as Key Targets for Vaccination and Therapeutic Interventions. Vaccines (Basel) 2025; 13:330. [PMID: 40266235 PMCID: PMC11946325 DOI: 10.3390/vaccines13030330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/12/2025] [Accepted: 03/17/2025] [Indexed: 04/24/2025] Open
Abstract
CD169 is a sialic acid-binding immunoglobulin-like lectin (Siglec-1, sialoadhesin) that is expressed by subsets of tissue-resident macrophages and circulating monocytes. This receptor interacts with α2,3-linked Neu5Ac on glycoproteins as well as glycolipids present on the surface of immune cells and pathogens. CD169-expressing macrophages exert tissue-specific homeostatic functions, but they also have opposing effects on the immune response. CD169+ macrophages act as a pathogen filter, protect against infectious diseases, and enhance adaptive immunity, but at the same time pathogens also exploit them to enable further dissemination. In cancer, CD169+ macrophages in tumor-draining lymph nodes are correlated with better clinical outcomes. In inflammatory diseases, CD169 expression is upregulated on monocytes and on monocyte-derived macrophages and this correlates with the disease state. Given their role in promoting adaptive immunity, CD169+ macrophages are currently investigated as targets for vaccination strategies against cancer. In this review, we describe the studies investigating the importance of CD169 and CD169+ macrophages in several disease settings and the vaccination strategies currently under investigation.
Collapse
Affiliation(s)
- Rianne G. Bouma
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Cancer Immunology, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
| | - Aru Z. Wang
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Cancer Immunology, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
| | - Joke M. M. den Haan
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Cancer Immunology, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
7
|
An H, Huang Y, Zhao Z, Li K, Meng J, Huang X, Tian X, Zhou H, Wu J, Dai Q, Zhang JR. Splenic red pulp macrophages eliminate the liver-resistant Streptococcus pneumoniae from the blood circulation of mice. SCIENCE ADVANCES 2025; 11:eadq6399. [PMID: 40073120 PMCID: PMC11900858 DOI: 10.1126/sciadv.adq6399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025]
Abstract
Invasive infections by encapsulated bacteria are the major cause of human morbidity and mortality. The liver resident macrophages, Kupffer cells, form the hepatic firewall to clear many encapsulated bacteria in the blood circulation but fail to control certain high-virulence capsule types. Here we report that the spleen is the backup immune organ to clear the liver-resistant serotypes of Streptococcus pneumoniae (pneumococcus), a leading human pathogen. Asplenic mice failed to control the growth of the liver-resistant pneumococci in the blood circulation. Immunologic and genetic analyses identified splenic red pulp (RP) macrophages as the major phagocytes for bacterial clearance. Furthermore, the plasma natural antibodies against the cell wall phosphocholine and the complement system were necessary for RP macrophage-mediated immunity. These findings have provided a conceptual framework for the innate defense against blood bacterial infections, a mechanistic explanation for the hyper-susceptibility of asplenic individuals to S. pneumoniae, and a proof of concept for developing vaccines and therapeutic antibodies against encapsulated pathogens.
Collapse
Affiliation(s)
- Haoran An
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China
- Department of Microbiology and Infectious Disease Center, Peking University Health Science Center, Beijing 100191, China
- Center for Infectious Biology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Yijia Huang
- Department of Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhifeng Zhao
- Department of Automation, Tsinghua University, Beijing 100084, China
| | - Kunpeng Li
- Center for Infectious Biology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Jingjing Meng
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China
| | - Xueting Huang
- Center for Infectious Biology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Xianbin Tian
- Center for Infectious Biology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Hongyu Zhou
- Department of Automation, Tsinghua University, Beijing 100084, China
| | - Jiamin Wu
- Department of Automation, Tsinghua University, Beijing 100084, China
| | - Qionghai Dai
- Department of Automation, Tsinghua University, Beijing 100084, China
| | - Jing-Ren Zhang
- Center for Infectious Biology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
8
|
Pont S, Nilly F, Berry L, Bonhoure A, Alford MA, Louis M, Nogaret P, Bains M, Lesouhaitier O, Hancock REW, Plésiat P, Blanc-Potard AB. Intracellular Pseudomonas aeruginosa persist and evade antibiotic treatment in a wound infection model. PLoS Pathog 2025; 21:e1012922. [PMID: 39946497 PMCID: PMC11825101 DOI: 10.1371/journal.ppat.1012922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 01/22/2025] [Indexed: 02/17/2025] Open
Abstract
Persistent bacterial infections evade host immunity and resist antibiotic treatments through various mechanisms that are difficult to evaluate in a living host. Pseudomonas aeruginosa is a main cause of chronic infections in patients with cystic fibrosis (CF) and wounds. Here, by immersing wounded zebrafish embryos in a suspension of P. aeruginosa isolates from CF patients, we established a model of persistent infection that mimics a murine chronic skin infection model. Live and electron microscopy revealed persisting aggregated P. aeruginosa inside zebrafish cells, including macrophages, at unprecedented resolution. Persistent P. aeruginosa exhibited adaptive resistance to several antibiotics, host cell permeable drugs being the most efficient. Moreover, persistent bacteria could be partly re-sensitized to antibiotics upon addition of anti-biofilm molecules that dispersed the bacterial aggregates in vivo. Collectively, this study demonstrates that an intracellular location protects persistent P. aeruginosa in vivo in wounded zebrafish embryos from host innate immunity and antibiotics, and provides new insights into efficient treatments against chronic infections.
Collapse
Affiliation(s)
- Stéphane Pont
- Laboratory of Pathogens and Host Immunity (LPHI), Université de Montpellier, CNRS, Inserm, 34095, Montpellier, France
| | - Flore Nilly
- Laboratory of Pathogens and Host Immunity (LPHI), Université de Montpellier, CNRS, Inserm, 34095, Montpellier, France
| | - Laurence Berry
- Laboratory of Pathogens and Host Immunity (LPHI), Université de Montpellier, CNRS, Inserm, 34095, Montpellier, France
| | - Anne Bonhoure
- Laboratory of Pathogens and Host Immunity (LPHI), Université de Montpellier, CNRS, Inserm, 34095, Montpellier, France
| | - Morgan A. Alford
- Center for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, Canada
| | - Mélissande Louis
- CBSA UR4312, Laboratoire de microbiologie Communication Bactérienne et Stratégies Anti-Infectieuses, Univ Rouen Normandie, Université Caen Normandie, Normandie Univ, Rouen, France
| | - Pauline Nogaret
- Laboratory of Pathogens and Host Immunity (LPHI), Université de Montpellier, CNRS, Inserm, 34095, Montpellier, France
| | - Manjeet Bains
- Center for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, Canada
| | - Olivier Lesouhaitier
- CBSA UR4312, Laboratoire de microbiologie Communication Bactérienne et Stratégies Anti-Infectieuses, Univ Rouen Normandie, Université Caen Normandie, Normandie Univ, Rouen, France
| | - Robert E. W. Hancock
- Center for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, Canada
| | - Patrick Plésiat
- UMR6249 CNRS Chrono-environnement, Université de Franche-Comté, Besançon, France
| | - Anne-Béatrice Blanc-Potard
- Laboratory of Pathogens and Host Immunity (LPHI), Université de Montpellier, CNRS, Inserm, 34095, Montpellier, France
| |
Collapse
|
9
|
Tian JH, Huang S, Wang ZH, Li JJ, Song X, Jiang ZT, Shi BS, Zhao YY, Zhang HY, Wang KR, Hu XY, Zhang X, Guo DS. Supramolecular discrimination and diagnosis-guided treatment of intracellular bacteria. Nat Commun 2025; 16:1016. [PMID: 39863571 PMCID: PMC11762306 DOI: 10.1038/s41467-025-56308-9] [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: 06/28/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Pathogenic intracellular bacteria pose a significant threat to global public health due to the barriers presented by host cells hindering the timely detection of hidden bacteria and the effective delivery of therapeutic agents. To address these challenges, we propose a tandem diagnosis-guided treatment paradigm. A supramolecular sensor array is developed for simple, rapid, accurate, and high-throughput identification of intracellular bacteria. This diagnostic approach executes the significant guiding missions of screening a customized host-guest drug delivery system by disclosing the rationale behind the discrimination. We design eight azocalix[4]arenes with differential active targeting, cellular internalization, and hypoxia responsiveness to penetrate cells and interact with bacteria. Loaded with fluorescent indicators, these azocalix[4]arenes form a sensor array capable of discriminating eight intracellular bacterial species without cell lysis or separation. By fingerprinting specimens collected from bacteria-infected mice, the facilitated accurate diagnosis offers valuable guidance for selecting appropriate antibiotics. Moreover, mannose-modified azocalix[4]arene (ManAC4A) is screened as a drug carrier efficiently taken up by macrophages. Doxycycline loaded with ManAC4A exhibits improved efficacy against methicillin-resistant Staphylococcus aureus-infected peritonitis. This study introduces an emerging paradigm to intracellular bacterial diagnosis and treatment, offering broad potential in combating bacterial infectious diseases.
Collapse
Affiliation(s)
- Jia-Hong Tian
- College of Chemistry, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China
| | - Siyuan Huang
- College of Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Tianjin Key Laboratory of Functional Polymer Materials, Nankai University, Tianjin, China
| | - Ze-Han Wang
- College of Chemistry, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China
| | - Juan-Juan Li
- College of Chemistry, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China
| | - Xianhui Song
- College of Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Tianjin Key Laboratory of Functional Polymer Materials, Nankai University, Tianjin, China
| | - Ze-Tao Jiang
- College of Chemistry, Nankai University, Tianjin, China
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China
| | - Bing-Sen Shi
- College of Chemistry and Materials Science, Hebei University, Baoding, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Hebei University, Baoding, China
- Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, China
- Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding, China
| | - Ying-Ying Zhao
- College of Chemistry and Materials Science, Hebei University, Baoding, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Hebei University, Baoding, China
- Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, China
- Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding, China
| | - Hui-Yan Zhang
- College of Chemistry and Materials Science, Hebei University, Baoding, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Hebei University, Baoding, China
- Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, China
- Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding, China
| | - Ke-Rang Wang
- College of Chemistry and Materials Science, Hebei University, Baoding, China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, China
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), Hebei University, Baoding, China
- Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, China
- Hebei Research Center of the Basic Discipline of Synthetic Chemistry, Hebei University, Baoding, China
| | - Xin-Yue Hu
- College of Chemistry, Nankai University, Tianjin, China.
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China.
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China.
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China.
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China.
| | - Xinge Zhang
- College of Chemistry, Nankai University, Tianjin, China.
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China.
- Tianjin Key Laboratory of Functional Polymer Materials, Nankai University, Tianjin, China.
| | - Dong-Sheng Guo
- College of Chemistry, Nankai University, Tianjin, China.
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin, China.
- Key Laboratory of Functional Polymer Materials (Ministry of Education), Nankai University, Tianjin, China.
- Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, China.
- Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, China.
- Xinjiang Key Laboratory of Novel Functional Materials Chemistry, Kashi University, Kashi, China.
- College of Chemistry and Environmental Sciences, Kashi University, Kashi, China.
| |
Collapse
|
10
|
Li W, Ding Q, Li M, Zhang T, Li C, Qi M, Dong B, Fang J, Wang L, Kim JS. Stimuli-responsive and targeted nanomaterials: Revolutionizing the treatment of bacterial infections. J Control Release 2025; 377:495-523. [PMID: 39580080 DOI: 10.1016/j.jconrel.2024.11.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/13/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024]
Abstract
Bacterial infections have emerged as a major threat to global public health. The effectiveness of traditional antibiotic treatments is waning due to the increasing prevalence of antimicrobial resistance, leading to an urgent demand for alternative antibacterial technologies. In this context, antibacterial nanomaterials have proven to be powerful tools for treating antibiotic-resistant and recurring infections. Targeting nanomaterials not only enable the precise delivery of bactericidal agents but also ensure controlled release at the infection site, thereby reducing potential systemic side effects. This review collates and categorizes nanomaterial-based responsive and precision-targeted antibacterial strategies into three key types: exogenous stimuli-responsive (including light, ultrasound, magnetism), bacterial microenvironment-responsive (such as pH, enzymes, hypoxia), and targeted antibacterial action (involving electrostatic interaction, covalent bonding, receptor-ligand mechanisms). Furthermore, we discuss recent advances, potential mechanisms, and future prospects in responsive and targeted antimicrobial nanomaterials, aiming to provide a comprehensive overview of the field's development and inspire the formulation of novel, precision-targeted antimicrobial strategies.
Collapse
Affiliation(s)
- Wen Li
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China; Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Qihang Ding
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China; Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Meiqi Li
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Tianshou Zhang
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Chunyan Li
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Manlin Qi
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Biao Dong
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130021, China.
| | - Jiao Fang
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China.
| | - Lin Wang
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China; Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China.
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
11
|
Wang ZJ, Zhu YY, Bai LY, Tang DM, Zhou ZS, Wei MZ, He JB, Yu-Duan, Luo XD. A new therapeutic strategy for infectious diseases against intracellular multidrug-resistant bacteria. J Control Release 2024; 375:467-477. [PMID: 39293527 DOI: 10.1016/j.jconrel.2024.09.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/09/2024] [Accepted: 09/14/2024] [Indexed: 09/20/2024]
Abstract
Bacterial infections result in 7,700,000 deaths per year globally, with intracellular bacteria causing repeated and resistant infection. No drug is currently licenced for the treatment of intracellular bacteria. A new screening platform mimicking the host milieu has been established to explore phytochemical antibiotic adjuvants. Previously neglected isoprenylated flavonoids were found to be effective against methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant enterococci (VRE). Specifically, the synergistic effect between glabrol and streptomycin against intracellular bacteria was observed for the first time. The glabrol-streptomycin combination targets bacterial inner membrane phospholipids, disrupts arginine biosynthesis, inhibits cell wall proteins and biofilm formation genes (agrA/B/C/D), and promotes ROS production, causing subsequent membrane and wall damage. To enhance the selective uptake of combination drug into infected cells, hyaluronic acid-streptomycin-lipoic acid-glabrol nanoparticles (HSLGS-S) were designed and synthesized to trigger the intracellular delivery of the glabrol-streptomycin combination. Thus, the treatment can be transported into the infected intracellular region and selectively release the glabrol-streptomycin combination to the bacterial at site. The bioactivity of HSLGS-S in clearing intracellular bacteria was 20-fold higher than that of the glabrol-streptomycin combination alone in vitro and 2- to 10-fold higher in vivo.
Collapse
Affiliation(s)
- Zhao-Jie Wang
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, PR China
| | - Yan-Yan Zhu
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, PR China
| | - Li-Yu Bai
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, PR China
| | - Dong-Mei Tang
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, PR China
| | - Zhong-Shun Zhou
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, PR China
| | - Mei-Zhen Wei
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, PR China
| | - Jin-Biao He
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, PR China
| | - Yu-Duan
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, PR China
| | - Xiao-Dong Luo
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming 650500, PR China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, PR China.
| |
Collapse
|
12
|
Urban BC, Gonçalves ANA, Loukov D, Passos FM, Reiné J, Gonzalez-Dias P, Solórzano C, Mitsi E, Nikolaou E, O'Connor D, Collins AM, Adler H, Pollard A, Rylance J, Gordon SB, Jochems SP, Nakaya HI, Ferreira DM. Inflammation of the nasal mucosa is associated with susceptibility to experimental pneumococcal challenge in older adults. Mucosal Immunol 2024; 17:973-989. [PMID: 38950826 PMCID: PMC11464406 DOI: 10.1016/j.mucimm.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024]
Abstract
Streptococcus pneumoniae colonization in the upper respiratory tract is linked to pneumococcal disease development, predominantly affecting young children and older adults. As the global population ages and comorbidities increase, there is a heightened concern about this infection. We investigated the immunological responses of older adults to pneumococcal-controlled human infection by analyzing the cellular composition and gene expression in the nasal mucosa. Our comparative analysis with data from a concurrent study in younger adults revealed distinct gene expression patterns in older individuals susceptible to colonization, highlighted by neutrophil activation and elevated levels of CXCL9 and CXCL10. Unlike younger adults challenged with pneumococcus, older adults did not show recruitment of monocytes into the nasal mucosa following nasal colonization. However, older adults who were protected from colonization showed increased degranulation of cluster of differentiation 8+ T cells, both before and after pneumococcal challenge. These findings suggest age-associated cellular changes, in particular enhanced mucosal inflammation, that may predispose older adults to pneumococcal colonization.
Collapse
Affiliation(s)
- Britta C Urban
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - André N A Gonçalves
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Dessi Loukov
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Fernando M Passos
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Jesús Reiné
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Patrícia Gonzalez-Dias
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Carla Solórzano
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Elena Mitsi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Elissavet Nikolaou
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; Infection, Immunity and Global Health, Murdoch Children's Research Institute, Parkville, Victoria, Australia; Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, Australia
| | - Daniel O'Connor
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Andrea M Collins
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; University Hospital Aintree, Liverpool University Hospitals Trust, Liverpool, UK
| | - Hugh Adler
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Andrew Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Jamie Rylance
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Stephen B Gordon
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; Malawi-Liverpool-Wellcome Clinical Research Programme, Blantyre, Malawi
| | - Simon P Jochems
- Leiden University Centre for Infectious Diseases, Leiden University Medical Centre, Leiden, The Netherlands
| | - Helder I Nakaya
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Daniela M Ferreira
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK.
| |
Collapse
|
13
|
Waltmann A, Balthazar JT, Begum AA, Hua N, Jerse AE, Shafer WM, Hobbs MM, Duncan JA. Experimental genital tract infection demonstrates Neisseria gonorrhoeae MtrCDE efflux pump is not required for in vivo human infection and identifies gonococcal colonization bottleneck. PLoS Pathog 2024; 20:e1012578. [PMID: 39321205 PMCID: PMC11457995 DOI: 10.1371/journal.ppat.1012578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 10/07/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024] Open
Abstract
The MtrCDE efflux pump of Neisseria gonorrhoeae exports a wide range of antimicrobial compounds that the gonococcus encounters at mucosal surfaces during colonization and infection and is a known gonococcal virulence factor. Here, we evaluate the role of this efflux pump system in strain FA1090 during in vivo human male urethral infection with N. gonorrhoeae using a controlled human infection model. With the strategy of competitive infections initiated with mixtures of wild-type FA1090 and an isogenic mutant FA1090 strain that does not contain a functional MtrCDE pump, we found that the presence of the efflux pump is not required for an infection to be established in the human male urethra. This finding contrasts with previous studies of in vivo infection in the lower genital tract of female mice, which demonstrated that mutant gonococci of a different strain (FA19) lacking a functional MtrCDE pump had a significantly reduced fitness compared to their wild-type parental FA19 strain. To determine if these conflicting results are due to strain or human vs. mouse differences, we conducted a series of systematic competitive infections in female mice with the same FA1090 strains as in humans, and with FA19 strains, including mutants that do not assemble a functional MtrCDE efflux pump. Our results indicate the fitness advantage provided by the MtrCDE efflux pump during infection of mice is strain dependent. Owing to the equal fitness of the two FA1090 strains in men, our experiments also demonstrated the presence of a colonization bottleneck of N. gonorrhoeae in the human male urethra, which may open a new area of inquiry into N. gonorrhoeae infection dynamics and control. TRIAL REGISTRATION. Clinicaltrials.gov NCT03840811.
Collapse
Affiliation(s)
- Andreea Waltmann
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States
| | - Jacqueline T. Balthazar
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Afrin A. Begum
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, Maryland, United States
| | - Nancy Hua
- The Emmes Company, Rockville, Maryland, United States
| | - Ann E. Jerse
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, Maryland, United States
| | - William M. Shafer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States
- The Emory Antibiotic Resistance Center, Emory University School of Medicine, Atlanta, Georgia, United States
- Laboratories of Bacterial Pathogenesis, Veterans Affairs Medical Center (Atlanta), Decatur, Georgia, United States
| | - Marcia M. Hobbs
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Joseph A. Duncan
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States
| |
Collapse
|
14
|
Benucci B, Spinello Z, Calvaresi V, Viviani V, Perrotta A, Faleri A, Utrio Lanfaloni S, Pansegrau W, d’Alterio L, Bartolini E, Pinzuti I, Sampieri K, Giordano A, Rappuoli R, Pizza M, Masignani V, Norais N, Maione D, Merola M. Neisserial adhesin A (NadA) binds human Siglec-5 and Siglec-14 with high affinity and promotes bacterial adhesion/invasion. mBio 2024; 15:e0110724. [PMID: 39041817 PMCID: PMC11323535 DOI: 10.1128/mbio.01107-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 05/20/2024] [Indexed: 07/24/2024] Open
Abstract
Neisserial adhesin A (NadA) is a meningococcal surface protein included as recombinant antigen in 4CMenB, a protein-based vaccine able to induce protective immune responses against Neisseria meningitidis serogroup B (MenB). Although NadA is involved in the adhesion/invasion of epithelial cells and human myeloid cells, its function in meningococcal physiology is still poorly understood. To clarify the role played by NadA in the host-pathogen interaction, we sought to identify its cellular receptors. We screened a protein microarray encompassing 2,846 human and 297 mouse surface/secreted recombinant proteins using recombinant NadA as probe. Efficient NadA binding was revealed on the paired sialic acid-binding immunoglobulin-type lectins receptors 5 and 14 (Siglec-5 and Siglec-14), but not on Siglec-9 therein used as control. The interaction was confirmed by biochemical tools with the determination of the KD value in the order of nanomolar and the identification of the NadA binding site by hydrogen-deuterium exchange coupled to mass spectrometry. The N-terminal domain of the Siglec-5 that recognizes the sialic acid was identified as the NadA binding domain. Intriguingly, exogenously added recombinant soluble Siglecs, including Siglec-9, were found to decorate N. meningitidis surface in a NadA-dependent manner. However, Siglec-5 and Siglec-14 transiently expressed in CHO-K1 cells endorsed NadA binding and increased N. meningitidis adhesion/invasion while Siglec-9 did not. Taken together, Siglec-5 and Siglec-14 satisfy all features of NadA receptors suggesting a possible role of NadA in the acute meningococcal infection.IMPORTANCEBacteria have developed several strategies for cell colonization and immune evasion. Knowledge of the host and pathogen factors involved in these mechanisms is crucial to build efficacious countermoves. Neisserial adhesin A (NadA) is a meningococcal surface protein included in the anti-meningococcus B vaccine 4CMenB, which mediates adhesion to and invasion of epithelial cells. Although NadA has been shown to bind to other cell types, like myeloid and endothelial cells, it still remains orphan of a defined host receptor. We have identified two strong NadA interactors, Siglec-5 and Siglec-14, which are mainly expressed on myeloid cells. This showcases that NadA is an additional and key player among the Neisseria meningitidis factors targeting immune cells. We thus provide novel insights on the strategies exploited by N. meningitidis during the infection process, which can progress to a severe illness and death.
Collapse
MESH Headings
- Humans
- Adhesins, Bacterial/metabolism
- Adhesins, Bacterial/genetics
- Bacterial Adhesion
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Lectins/metabolism
- Lectins/genetics
- Lectins/immunology
- Animals
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Host-Pathogen Interactions
- Protein Binding
- Mice
- CHO Cells
- Cricetulus
- Neisseria meningitidis/genetics
- Neisseria meningitidis/metabolism
- Neisseria meningitidis/immunology
- Recombinant Proteins/metabolism
- Recombinant Proteins/genetics
- Sialic Acid Binding Immunoglobulin-like Lectins/metabolism
- Sialic Acid Binding Immunoglobulin-like Lectins/genetics
- Epithelial Cells/microbiology
- Epithelial Cells/metabolism
- Epithelial Cells/immunology
- Meningococcal Infections/microbiology
- Meningococcal Infections/immunology
- Receptors, Cell Surface/metabolism
- Receptors, Cell Surface/genetics
- Neisseria meningitidis, Serogroup B/genetics
- Neisseria meningitidis, Serogroup B/immunology
- Neisseria meningitidis, Serogroup B/metabolism
Collapse
Affiliation(s)
| | | | - Valeria Calvaresi
- GSK, Siena, Italy
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Marcello Merola
- GSK, Siena, Italy
- Università di Napoli Federico II, Naples, Italy
| |
Collapse
|
15
|
Hernandez-Morfa M, Reinoso-Vizcaino NM, Zappia VE, Olivero NB, Cortes PR, Stempin CC, Perez DR, Echenique J. Intracellular Streptococcus pneumoniae develops enhanced fluoroquinolone persistence during influenza A coinfection. Front Microbiol 2024; 15:1423995. [PMID: 39035445 PMCID: PMC11258013 DOI: 10.3389/fmicb.2024.1423995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/18/2024] [Indexed: 07/23/2024] Open
Abstract
Streptococcus pneumoniae is a major pathogen responsible for severe complications in patients with prior influenza A virus (IAV) infection. We have previously demonstrated that S. pneumoniae exhibits increased intracellular survival within IAV-infected cells. Fluoroquinolones (FQs) are widely used to treat pneumococcal infections. However, our prior work has shown that S. pneumoniae can develop intracellular FQ persistence, a phenomenon triggered by oxidative stress within host cells. This persistence allows the bacteria to withstand high FQ concentrations. In this study, we show that IAV infection enhances pneumococcal FQ persistence during intracellular survival within pneumocytes, macrophages, and neutrophils. This enhancement is partly due to increased oxidative stress induced by the viral infection. We find that this phenotype is particularly pronounced in autophagy-proficient host cells, potentially resulting from IAV-induced blockage of autophagosome-lysosome fusion. Moreover, we identified several S. pneumoniae genes involved in oxidative stress response that contribute to FQ persistence, including sodA (superoxide dismutase), clpL (chaperone), nrdH (glutaredoxin), and psaB (Mn+2 transporter component). Our findings reveal a novel mechanism of antibiotic persistence promoted by viral infection within host cells. This underscores the importance of considering this phenomenon when using FQs to treat pneumococcal infections, especially in patients with concurrent influenza A infection.
Collapse
Affiliation(s)
- Mirelys Hernandez-Morfa
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Nicolas M. Reinoso-Vizcaino
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Victoria E. Zappia
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Nadia B. Olivero
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Paulo R. Cortes
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Cinthia C. Stempin
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Daniel R. Perez
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Jose Echenique
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
16
|
Korkmaz FT, Quinton LJ. Extra-pulmonary control of respiratory defense. Cell Immunol 2024; 401-402:104841. [PMID: 38878619 PMCID: PMC12002097 DOI: 10.1016/j.cellimm.2024.104841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 06/06/2024] [Indexed: 07/13/2024]
Abstract
Pneumonia persists as a public health crisis, representing the leading cause of death due to infection. Whether respiratory tract infections progress to pneumonia and its sequelae such as acute respiratory distress syndrome and sepsis depends on numerous underlying conditions related to both the causative agent and host. Regarding the former, pneumonia burden remains staggeringly high, despite the effectiveness of pathogen-targeting strategies such as vaccines and antibiotics. This demands a greater understanding of host features that collaborate to promote immune resistance and tissue resilience in the infected lung. Such features inside the pulmonary compartment have drawn much attention, where major advances have been made related to resident and recruited immune activity. By comparison, extra-pulmonary processes guiding pneumonia susceptibility are relatively elusive, constituting the focus of this review. Here we will highlight examples of when, how, and why tissues outside of the lungs dispatch signals that modulate local immunity in the airspaces. Topics include the liver, gut, bone marrow, brain and more, all of which contribute in direct and indirect ways to pneumonia outcome. When tuned appropriately, it has become clear that these responses can serve protective roles, and this will be considered distinctly from what would otherwise be aberrant responses characteristic of pneumonia-induced organ injury and sepsis. Further advances in this area may reveal novel targetable areas for clinical intervention that are not confined to the intra-pulmonary space.
Collapse
Affiliation(s)
- Filiz T Korkmaz
- Department of Medicine, Division of Immunology and Infectious Disease, UMass Chan Medical School, Worcester, MA 01602, United States.
| | - Lee J Quinton
- Department of Medicine, Division of Immunology and Infectious Disease, UMass Chan Medical School, Worcester, MA 01602, United States
| |
Collapse
|
17
|
Li F, Zhu P, Zheng B, Lu Z, Fang C, Fu Y, Li X. A Customized Biohybrid Presenting Cascade Responses to Tumor Microenvironment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404901. [PMID: 38723206 DOI: 10.1002/adma.202404901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Indexed: 05/16/2024]
Abstract
Intrinsic characteristics of microorganisms, including non-specific metabolism sites, toxic byproducts, and uncontrolled proliferation constrain their exploitation in medical applications such as tumor therapy. Here, the authors report an engineered biohybrid that can efficiently target cancerous sites through a pre-determined metabolic pathway to enable precise tumor ablation. In this system, DH5α Escherichia coli is engineered by the introduction of hypoxia-inducible promoters and lactate oxidase genes, and further surface-armored with iron-doped ZIF-8 nanoparticles. This bioengineered E. coli can produce and secrete lactate oxidase to reduce lactate concentration in response to hypoxic tumor microenvironment, as well as triggering immune activation. The peroxidase-like functionality of the nanoparticles extends the end product of the lactate metabolism, enabling the conversion of hydrogen peroxide (H2O2) into highly cytotoxic hydroxyl radicals. This, coupled with the transformation of tirapazamine loaded on nanoparticles to toxic benzotriazinyl, culminates in severe tumor cell ferroptosis. Intravenous injection of this biohybrid significantly inhibits tumor growth and metastasis.
Collapse
Affiliation(s)
- Feiyu Li
- State Key Laboratory of Silicon and Advanced Semiconductor Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310058, China
- ZJU-Hangzhou Global Science and Technology Innovation Center, Zhejiang University, Hangzhou, 311215, China
| | - Peipei Zhu
- State Key Laboratory of Silicon and Advanced Semiconductor Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310058, China
- ZJU-Hangzhou Global Science and Technology Innovation Center, Zhejiang University, Hangzhou, 311215, China
| | - Bingzhu Zheng
- State Key Laboratory of Silicon and Advanced Semiconductor Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310058, China
- ZJU-Hangzhou Global Science and Technology Innovation Center, Zhejiang University, Hangzhou, 311215, China
| | - Zijie Lu
- State Key Laboratory of Silicon and Advanced Semiconductor Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310058, China
- ZJU-Hangzhou Global Science and Technology Innovation Center, Zhejiang University, Hangzhou, 311215, China
| | - Chao Fang
- State Key Laboratory of Silicon and Advanced Semiconductor Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310058, China
- ZJU-Hangzhou Global Science and Technology Innovation Center, Zhejiang University, Hangzhou, 311215, China
| | - Yike Fu
- State Key Laboratory of Silicon and Advanced Semiconductor Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310058, China
- ZJU-Hangzhou Global Science and Technology Innovation Center, Zhejiang University, Hangzhou, 311215, China
| | - Xiang Li
- State Key Laboratory of Silicon and Advanced Semiconductor Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310058, China
- ZJU-Hangzhou Global Science and Technology Innovation Center, Zhejiang University, Hangzhou, 311215, China
| |
Collapse
|
18
|
Arron HE, Marsh BD, Kell DB, Khan MA, Jaeger BR, Pretorius E. Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: the biology of a neglected disease. Front Immunol 2024; 15:1386607. [PMID: 38887284 PMCID: PMC11180809 DOI: 10.3389/fimmu.2024.1386607] [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: 02/15/2024] [Accepted: 04/11/2024] [Indexed: 06/20/2024] Open
Abstract
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a chronic, debilitating disease characterised by a wide range of symptoms that severely impact all aspects of life. Despite its significant prevalence, ME/CFS remains one of the most understudied and misunderstood conditions in modern medicine. ME/CFS lacks standardised diagnostic criteria owing to variations in both inclusion and exclusion criteria across different diagnostic guidelines, and furthermore, there are currently no effective treatments available. Moving beyond the traditional fragmented perspectives that have limited our understanding and management of the disease, our analysis of current information on ME/CFS represents a significant paradigm shift by synthesising the disease's multifactorial origins into a cohesive model. We discuss how ME/CFS emerges from an intricate web of genetic vulnerabilities and environmental triggers, notably viral infections, leading to a complex series of pathological responses including immune dysregulation, chronic inflammation, gut dysbiosis, and metabolic disturbances. This comprehensive model not only advances our understanding of ME/CFS's pathophysiology but also opens new avenues for research and potential therapeutic strategies. By integrating these disparate elements, our work emphasises the necessity of a holistic approach to diagnosing, researching, and treating ME/CFS, urging the scientific community to reconsider the disease's complexity and the multifaceted approach required for its study and management.
Collapse
Affiliation(s)
- Hayley E. Arron
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Benjamin D. Marsh
- MRCPCH Consultant Paediatric Neurodisability, Exeter, Devon, United Kingdom
| | - Douglas B. Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - M. Asad Khan
- Directorate of Respiratory Medicine, Manchester University Hospitals, Wythenshawe Hospital, Manchester, United Kingdom
| | - Beate R. Jaeger
- Long COVID department, Clinic St Georg, Bad Aibling, Germany
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
19
|
Lokken-Toyli KL, Aggarwal SD, Bee GCW, de Steenhuijsen Piters WAA, Wu C, Chen KZM, Loomis C, Bogaert D, Weiser JN. Impaired upper respiratory tract barrier function during postnatal development predisposes to invasive pneumococcal disease. PLoS Pathog 2024; 20:e1012111. [PMID: 38718049 PMCID: PMC11078396 DOI: 10.1371/journal.ppat.1012111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/10/2024] [Indexed: 05/12/2024] Open
Abstract
Infants are highly susceptible to invasive respiratory and gastrointestinal infections. To elucidate the age-dependent mechanism(s) that drive bacterial spread from the mucosa, we developed an infant mouse model using the prevalent pediatric respiratory pathogen, Streptococcus pneumoniae (Spn). Despite similar upper respiratory tract (URT) colonization levels, the survival rate of Spn-infected infant mice was significantly decreased compared to adults and corresponded with Spn dissemination to the bloodstream. An increased rate of pneumococcal bacteremia in early life beyond the newborn period was attributed to increased bacterial translocation across the URT barrier. Bacterial dissemination in infant mice was independent of URT monocyte or neutrophil infiltration, phagocyte-derived ROS or RNS, inflammation mediated by toll-like receptor 2 or interleukin 1 receptor signaling, or the pore-forming toxin pneumolysin. Using molecular barcoding of Spn, we found that only a minority of bacterial clones in the nasopharynx disseminated to the blood in infant mice, indicating the absence of robust URT barrier breakdown. Rather, transcriptional profiling of the URT epithelium revealed a failure of infant mice to upregulate genes involved in the tight junction pathway. Expression of many such genes was also decreased in early life in humans. Infant mice also showed increased URT barrier permeability and delayed mucociliary clearance during the first two weeks of life, which corresponded with tighter attachment of bacteria to the respiratory epithelium. Together, these results demonstrate a window of vulnerability during postnatal development when altered mucosal barrier function facilitates bacterial dissemination.
Collapse
Affiliation(s)
- Kristen L. Lokken-Toyli
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
| | - Surya D. Aggarwal
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
| | - Gavyn Chern Wei Bee
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
| | - Wouter A. A. de Steenhuijsen Piters
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children’s Hospital/University Medical Center Utrecht, Utrecht, the Netherlands
- National Institute for Public Health and the Environment, Bilthoven, the Netherlands; Department of Parasitology, Leiden University Medical Center, Leiden, the Netherlands
| | - Cindy Wu
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
| | - Kenny Zhi Ming Chen
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
| | - Cynthia Loomis
- Department of Pathology, New York University School of Medicine, New York, New York, United States of America
| | - Debby Bogaert
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children’s Hospital/University Medical Center Utrecht, Utrecht, the Netherlands
- Centre for Inflammation Research, Institute for Regeneration and Repair, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Jeffrey N. Weiser
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
| |
Collapse
|
20
|
Xia Z, Liao Y, Gao G, Zhang S. Rifampicin-Loaded Polyelectrolyte Complex Eliminates Intracellular Bacteria through Thiol-Mediated Cellular Uptake and Oxidative Stress Enhancement. ACS APPLIED BIO MATERIALS 2024; 7:2544-2553. [PMID: 38507285 DOI: 10.1021/acsabm.4c00143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
The poor accumulation of antibiotics in the cytoplasm leads to the poor eradication of intracellular bacteria. Herein, a polyelectrolyte complex (PECs@Rif) allowing direct cytosolic delivery of rifampicin (Rif) was developed for the treatment of intracellular infections by complexation of poly(α-lipoic acid) (pLA) and oligosaccharide (COS) in water and loading Rif. Due to the thiol-mediated cellular uptake, PECs@Rif delivered 3.9 times higher Rif into the cytoplasm than that of the free Rif during 8 h of incubation. After entering cells, PECs@Rif released Rif by dissociating pLA into dihydrolipoic acid (DHLA) in the presence of intracellular thioredoxin reductase (TrxR). Notably, DHLA could reduce endogenous Fe(III) to Fe(II) and provide a catalyst for the Fenton reaction to produce a large amount of reactive oxygen species (ROS), which would assist Rif in eradicating intracellular bacteria. In vitro assay showed that PECs@Rif reduced almost 2.8 orders of magnitude of intracellular bacteria, much higher than 0.7 orders of magnitude of free Rif. The bacteremia-bearing mouse models showed that PECs@Rif reduced bacterial levels in the liver, spleen, and kidney by 2.2, 3.7, and 2.3 orders of magnitude, respectively, much higher than free Rif in corresponding tissues. The direct cytosolic delivery in a thiol-mediated manner and enhanced oxidative stress proposed a feasible strategy for treating intracellular bacteria infection.
Collapse
Affiliation(s)
- Zhaoxin Xia
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Yulong Liao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Ge Gao
- College of Chemistry, Sichuan University, Chengdu 610064, China
| | - Shiyong Zhang
- College of Chemistry, Sichuan University, Chengdu 610064, China
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| |
Collapse
|
21
|
Jansson MK, Nguyen DT, Mikkat S, Warnke C, Janssen MB, Warnke P, Kreikemeyer B, Patenge N. Synthetic mRNA delivered to human cells leads to expression of Cpl-1 bacteriophage-endolysin with activity against Streptococcus pneumoniae. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102145. [PMID: 38435119 PMCID: PMC10907214 DOI: 10.1016/j.omtn.2024.102145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 02/06/2024] [Indexed: 03/05/2024]
Abstract
Endolysins are bacteriophage-encoded hydrolases that show high antibacterial activity and a narrow substrate spectrum. We hypothesize that an mRNA-based approach to endolysin therapy can overcome some challenges of conventional endolysin therapy, namely organ targeting and bioavailability. We show that synthetic mRNA applied to three human cell lines (HEK293T, A549, HepG2 cells) leads to expression and cytosolic accumulation of the Cpl-1 endolysin with activity against Streptococcus pneumoniae. Addition of a human lysozyme signal peptide sequence translocates the Cpl-1 to the endoplasmic reticulum leading to secretion (hlySP-sCpl-1). The pneumococcal killing effect of hlySP-sCpl-1 was enhanced by introduction of a point mutation to avoid N-linked-glycosylation. hlySP-sCpl-1N215D, collected from the culture supernatant of A549 cells 6 h post-transfection showed a significant killing effect and was active against nine pneumococcal strains. mRNA-based cytosolic Cpl-1 and secretory hlySP-sCpl-1N215D show potential for innovative treatment strategies against pneumococcal disease and, to our best knowledge, represent the first approach to mRNA-based endolysin therapy. We assume that many other bacterial pathogens could be targeted with this novel approach.
Collapse
Affiliation(s)
- Moritz K. Jansson
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Dat Tien Nguyen
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Stefan Mikkat
- Core Facility Proteome Analysis, University Medicine Rostock, Rostock, Germany
| | - Carolin Warnke
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Marc Benjamin Janssen
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Philipp Warnke
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Bernd Kreikemeyer
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| | - Nadja Patenge
- Institute of Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Rostock, Germany
| |
Collapse
|
22
|
Santra S, Nayak I, Paladhi A, Das D, Banerjee A. Estimates of differential toxin expression governing heterogeneous intracellular lifespans of Streptococcus pneumoniae. J Cell Sci 2024; 137:jcs260891. [PMID: 38411297 DOI: 10.1242/jcs.260891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/10/2024] [Indexed: 02/28/2024] Open
Abstract
Following invasion of the host cell, pore-forming toxins secreted by pathogens compromise vacuole integrity and expose the microbe to diverse intracellular defence mechanisms. However, the quantitative correlation between toxin expression levels and consequent pore dynamics, fostering the intracellular life of pathogens, remains largely unexplored. In this study, using Streptococcus pneumoniae and its secreted pore-forming toxin pneumolysin (Ply) as a model system, we explored various facets of host-pathogen interactions in the host cytosol. Using time-lapse fluorescence imaging, we monitored pore formation dynamics and lifespans of different pneumococcal subpopulations inside host cells. Based on experimental histograms of various event timescales such as pore formation time, vacuolar death or cytosolic escape time and total degradation time, we developed a mathematical model based on first-passage processes that could correlate the event timescales to intravacuolar toxin accumulation. This allowed us to estimate Ply production rate, burst size and threshold Ply quantities that trigger these outcomes. Collectively, we present a general method that illustrates a correlation between toxin expression levels and pore dynamics, dictating intracellular lifespans of pathogens.
Collapse
Affiliation(s)
- Shweta Santra
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| | - Indrani Nayak
- Department of Physics, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| | - Ankush Paladhi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| | - Dibyendu Das
- Department of Physics, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| | - Anirban Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| |
Collapse
|
23
|
Nanushaj D, Kono M, Sakatani H, Murakami D, Hotomi M. Nucleic acid sensing Toll-like receptors 3 and 9 play complementary roles in the development of bacteremia after nasal colonization associated with influenza co-infection. Exp Anim 2024; 73:50-60. [PMID: 37532523 PMCID: PMC10877144 DOI: 10.1538/expanim.23-0001] [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: 01/01/2023] [Accepted: 07/30/2023] [Indexed: 08/04/2023] Open
Abstract
Streptococcus pneumoniae can cause mortality in infant, elderly, and immunocompromised individuals owing to invasion of bacteria to the lungs, the brain, and the blood. In building strategies against invasive infections, it is important to achieve greater understanding of how the pneumococci are able to survive in the host. Toll-like receptors (TLRs), critically important components in the innate immune system, have roles in various stages of the development of infectious diseases. Endosomal TLRs recognize nucleic acids of the pathogen, but the impact on the pneumococcal diseases of immune responses from signaling them remains unclear. To investigate their role in nasal colonization and invasive disease with/without influenza co-infection, we established a mouse model of invasive pneumococcal diseases directly developing from nasal colonization. TLR9 KO mice had bacteremia more frequently than wildtype in the pneumococcal mono-infection model, while the occurrence of bacteremia was higher among TLR3 KO mice after infection with influenza in advance of pneumococcal inoculation. All TLR KO strains showed poorer survival than wildtype after the mice had bacteremia. The specific and protective role of TLR3 and TLR9 was shown in developing bacteremia with/without influenza co-infection respectively, and all nucleic sensing TLRs would contribute equally to protecting sepsis after bacteremia.
Collapse
Affiliation(s)
- Denisa Nanushaj
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Research Building 9F, 811-1 Kimiidera, Wakayama 641-8510, Japan
| | - Masamitsu Kono
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Research Building 9F, 811-1 Kimiidera, Wakayama 641-8510, Japan
| | - Hideki Sakatani
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Research Building 9F, 811-1 Kimiidera, Wakayama 641-8510, Japan
| | - Daichi Murakami
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Research Building 9F, 811-1 Kimiidera, Wakayama 641-8510, Japan
| | - Muneki Hotomi
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Research Building 9F, 811-1 Kimiidera, Wakayama 641-8510, Japan
| |
Collapse
|
24
|
Xiong J, Tang H, Sun L, Zhu J, Tao S, Luo J, Li J, Li J, Wu H, Yang J. A macrophage cell membrane-coated cascade-targeting photothermal nanosystem for combating intracellular bacterial infections. Acta Biomater 2024; 175:293-306. [PMID: 38159895 DOI: 10.1016/j.actbio.2023.12.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/05/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
Current antibacterial interventions encounter formidable challenges when confronting intracellular bacteria, attributable to their clustering within phagocytes, particularly macrophages, evading host immunity and resisting antibiotics. Herein, we have developed an intelligent cell membrane-based nanosystem, denoted as MM@DAu NPs, which seamlessly integrates cascade-targeting capabilities with controllable antibacterial functions for the precise elimination of intracellular bacteria. MM@DAu NPs feature a core comprising D-alanine-functionalized gold nanoparticles (DAu NPs) enveloped by a macrophage cell membrane (MM) coating. Upon administration, MM@DAu NPs harness the intrinsic homologous targeting ability of their macrophage membrane to infiltrate bacteria-infected macrophages. Upon internalization within these host cells, exposed DAu NPs from MM@DAu NPs selectively bind to intracellular bacteria through the bacteria-targeting agent, D-alanine present on DAu NPs. This intricate process establishes a cascade mechanism that efficiently targets intracellular bacteria. Upon exposure to near-infrared irradiation, the accumulated DAu NPs surrounding intracellular bacteria induce local hyperthermia, enabling precise clearance of intracellular bacteria. Further validation in animal models infected with the typical intracellular bacteria, Staphylococcus aureus, substantiates the exceptional cascade-targeting efficacy and photothermal antibacterial potential of MM@DAu NPs in vivo. Therefore, this integrated cell membrane-based cascade-targeting photothermal nanosystem offers a promising approach for conquering persistent intracellular infections without drug resistance risks. STATEMENT OF SIGNIFICANCE: Intracellular bacterial infections lead to treatment failures and relapses because intracellular bacteria could cluster within phagocytes, especially macrophages, evading the host immune system and resisting antibiotics. Herein, we have developed an intelligent cell membrane-based nanosystem MM@DAu NPs, which is designed to precisely eliminate intracellular bacteria through a controllable cascade-targeting photothermal antibacterial approach. MM@DAu NPs combine D-alanine-functionalized gold nanoparticles with a macrophage cell membrane coating. Upon administration, MM@DAu NPs harness the homologous targeting ability of macrophage membrane to infiltrate bacteria-infected macrophages. Upon internalization, exposed DAu NPs from MM@DAu NPs selectively bind to intracellular bacteria through the bacteria-targeting agent, enabling precise clearance of intracellular bacteria through local hyperthermia. This integrated cell membrane-based cascade-targeting photothermal nanosystem offers a promising avenue for conquering persistent intracellular infections without drug resistance risks.
Collapse
Affiliation(s)
- Jingdi Xiong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Haiqin Tang
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Lizhong Sun
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jieyu Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Siying Tao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jun Luo
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Jiyao Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jianshu Li
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Hongkun Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Jiaojiao Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
25
|
Zheng X, Xing Y, Sun K, Jin H, Zhao W, Yu F. Combination Therapy with Resveratrol and Celastrol Using Folic Acid-Functionalized Exosomes Enhances the Therapeutic Efficacy of Sepsis. Adv Healthc Mater 2023; 12:e2301325. [PMID: 37530416 DOI: 10.1002/adhm.202301325] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/30/2023] [Indexed: 08/03/2023]
Abstract
Overactivated macrophages are a prominent feature of many inflammatory and autoimmune diseases, including sepsis. Attention and regulation of macrophages activity is of great significance for sepsis treatment. Herein, this study shows that folic acid-functionalized exosomes accumulate in the lung of septic mice and specifically target inflammatory macrophages. Therefore, FA-functionalized exosomes co-loaded with resveratrol (an anti-inflammatory polyphenol) and celastrol (an immunosuppressive pentacyclic triterpenoid; FA-Exo/R+C), which exhibit powerful anti-inflammatory and immunosuppressive activities against LPS-stimulated macrophages in vitro by regulating NF-κB and ERK1/2 signaling pathways, are designed. Encouraged by these positive data, the efficacy of FA-Exo/R+C is systematically investigated in an LPS-induced mouse sepsis model. FA-Exo/R+C shows striking therapeutic benefits in terms of attenuated cytokine storm, reduced acute lung injury, and increased survival of septic mice by inhibiting the inflammation and proliferation of proinflammatory M1 macrophages. Importantly, multiple administrations of FA-Exo/R+C significantly enhance and prolong the protective effect, and resist rechallenge to LPS. Collectively, the strategy of co-delivering drugs combination through functionalized exosomes offers a new avenue for sepsis treatment.
Collapse
Affiliation(s)
- Xue Zheng
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Yujie Xing
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Ke Sun
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Hongzhen Jin
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Wei Zhao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Fan Yu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, College of Life Sciences, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, Tongyan Road, Haihe Education Park, Tianjin, 300350, China
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, 266071, China
| |
Collapse
|
26
|
Gao Y, Tian X, Zhang X, Milebe Nkoua GD, Chen F, Liu Y, Chai Y. The roles of tissue-resident macrophages in sepsis-associated organ dysfunction. Heliyon 2023; 9:e21391. [PMID: 38027963 PMCID: PMC10643296 DOI: 10.1016/j.heliyon.2023.e21391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/13/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Sepsis, a syndrome caused by a dysregulated host response to infection and characterized by life-threatening organ dysfunction, particularly septic shock and sepsis-associated organ dysfunction (SAOD), is a medical emergency associated with high morbidity, high mortality, and long-term sequelae. Tissue-resident macrophages (TRMs) are a subpopulation of macrophages derived primarily from yolk sac progenitors and fetal liver during embryogenesis, located primarily in non-lymphoid tissues in adulthood, capable of local self-renewal independent of hematopoiesis, and developmentally and functionally restricted to the non-lymphoid organs in which they reside. TRMs are the first line of defense against life-threatening conditions such as sepsis, tumor growth, traumatic-associated organ injury, and surgical-associated injury. In the context of sepsis, TRMs can be considered as angels or demons involved in organ injury. Our proposal is that sepsis, septic shock, and SAOD can be attenuated by modulating TRMs in different organs. This review summarizes the pathophysiological mechanisms of TRMs in different organs or tissues involved in the development and progression of sepsis.
Collapse
Affiliation(s)
- Yulei Gao
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, P. R. China
- Department of Emergency Medicine, China-Congo Friendship Hospital, Brazzaville, 999059, P. R. Congo
| | - Xin Tian
- Department of Medical Research, Beijing Qiansong Technology Development Company, Beijing, 100193, P. R. China
- Department of Medical Research, Sen Sho Ka Gi Company, Inba-gun, Chiba, 285-0905, Japan
| | - Xiang Zhang
- Department of Emergency Medicine, Rizhao People's Hospital of Shandong Province, Rizhao, 276825, P. R. China
| | | | - Fang Chen
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, P. R. China
| | - Yancun Liu
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, P. R. China
| | - Yanfen Chai
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, P. R. China
| |
Collapse
|
27
|
Hernandez-Morfa M, Olivero NB, Zappia VE, Piñas GE, Reinoso-Vizcaino NM, Cian MB, Nuñez-Fernandez M, Cortes PR, Echenique J. The oxidative stress response of Streptococcus pneumoniae: its contribution to both extracellular and intracellular survival. Front Microbiol 2023; 14:1269843. [PMID: 37789846 PMCID: PMC10543277 DOI: 10.3389/fmicb.2023.1269843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 08/28/2023] [Indexed: 10/05/2023] Open
Abstract
Streptococcus pneumoniae is a gram-positive, aerotolerant bacterium that naturally colonizes the human nasopharynx, but also causes invasive infections and is a major cause of morbidity and mortality worldwide. This pathogen produces high levels of H2O2 to eliminate other microorganisms that belong to the microbiota of the respiratory tract. However, it also induces an oxidative stress response to survive under this stressful condition. Furthermore, this self-defense mechanism is advantageous in tolerating oxidative stress imposed by the host's immune response. This review provides a comprehensive overview of the strategies employed by the pneumococcus to survive oxidative stress. These strategies encompass the utilization of H2O2 scavengers and thioredoxins, the adaptive response to antimicrobial host oxidants, the regulation of manganese and iron homeostasis, and the intricate regulatory networks that control the stress response. Here, we have also summarized less explored aspects such as the involvement of reparation systems and polyamine metabolism. A particular emphasis is put on the role of the oxidative stress response during the transient intracellular life of Streptococcus pneumoniae, including coinfection with influenza A and the induction of antibiotic persistence in host cells.
Collapse
Affiliation(s)
- Mirelys Hernandez-Morfa
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Nadia B. Olivero
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Victoria E. Zappia
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - German E. Piñas
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Nicolas M. Reinoso-Vizcaino
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Melina B. Cian
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mariana Nuñez-Fernandez
- Centro de Química Aplicada, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Paulo R. Cortes
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Jose Echenique
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
28
|
Xia X. Horizontal Gene Transfer and Drug Resistance Involving Mycobacterium tuberculosis. Antibiotics (Basel) 2023; 12:1367. [PMID: 37760664 PMCID: PMC10526031 DOI: 10.3390/antibiotics12091367] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/13/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) acquires drug resistance at a rate comparable to that of bacterial pathogens that replicate much faster and have a higher mutation rate. One explanation for this rapid acquisition of drug resistance in Mtb is that drug resistance may evolve in other fast-replicating mycobacteria and then be transferred to Mtb through horizontal gene transfer (HGT). This paper aims to address three questions. First, does HGT occur between Mtb and other mycobacterial species? Second, what genes after HGT tend to survive in the recipient genome? Third, does HGT contribute to antibiotic resistance in Mtb? I present a conceptual framework for detecting HGT and analyze 39 ribosomal protein genes, 23S and 16S ribosomal RNA genes, as well as several genes targeted by antibiotics against Mtb, from 43 genomes representing all major groups within Mycobacterium. I also included mgtC and the insertion sequence IS6110 that were previously reported to be involved in HGT. The insertion sequence IS6110 shows clearly that the Mtb complex participates in HGT. However, the horizontal transferability of genes depends on gene function, as was previously hypothesized. HGT is not observed in functionally important genes such as ribosomal protein genes, rRNA genes, and other genes chosen as drug targets. This pattern can be explained by differential selection against functionally important and unimportant genes after HGT. Functionally unimportant genes such as IS6110 are not strongly selected against, so HGT events involving such genes are visible. For functionally important genes, a horizontally transferred diverged homologue from a different species may not work as well as the native counterpart, so the HGT event involving such genes is strongly selected against and eliminated, rendering them invisible to us. In short, while HGT involving the Mtb complex occurs, antibiotic resistance in the Mtb complex arose from mutations in those drug-targeted genes within the Mtb complex and was not gained through HGT.
Collapse
Affiliation(s)
- Xuhua Xia
- Department of Biology, University of Ottawa, Ottawa, ON K1N 9A7, Canada; ; Tel.: +1-613-562-5718
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
29
|
Camarasa TMN, Torné J, Chevalier C, Rasid O, Hamon MA. Streptococcus pneumoniae drives specific and lasting Natural Killer cell memory. PLoS Pathog 2023; 19:e1011159. [PMID: 37486946 PMCID: PMC10399893 DOI: 10.1371/journal.ppat.1011159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 08/03/2023] [Accepted: 06/27/2023] [Indexed: 07/26/2023] Open
Abstract
NK cells are important mediators of innate immunity and play an essential role for host protection against infection, although their responses to bacteria are poorly understood. Recently NK cells were shown to display memory properties, as characterized by an epigenetic signature leading to a stronger secondary response. Although NK cell memory could be a promising mechanism to fight against infection, it has not been described upon bacterial infection. Using a mouse model, we reveal that NK cells develop specific and long-term memory following sub-lethal infection with the extracellular pathogen Streptococcus pneumoniae. Memory NK cells display intrinsic sensing and response to bacteria in vitro, in a manner that is enhanced post-bacterial infection. In addition, their transfer into naïve mice confers protection from lethal infection for at least 12 weeks. Interestingly, NK cells display enhanced cytotoxic molecule production upon secondary stimulation and their protective role is dependent on Perforin and independent of IFNγ. Thus, our study identifies a new role for NK cells during bacterial infection, opening the possibility to harness innate immune memory for therapeutic purposes.
Collapse
Affiliation(s)
- Tiphaine M. N. Camarasa
- Chromatin and Infection Unit, Institut Pasteur, Paris, France
- Université Paris Cité, 562 Bio Sorbonne Paris Cité, Paris, France
| | - Júlia Torné
- Chromatin and Infection Unit, Institut Pasteur, Paris, France
| | | | - Orhan Rasid
- Chromatin and Infection Unit, Institut Pasteur, Paris, France
| | | |
Collapse
|
30
|
Waltmann A, Balthazar JT, Begum AA, Hua N, Jerse AE, Shafer WM, Hobbs MM, Duncan JA. Neisseria gonorrhoeae MtrCDE Efflux Pump During In Vivo Experimental Genital Tract Infection in Men and Mice Reveals the Presence of Within-Host Colonization Bottleneck. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.23.23291824. [PMID: 37425726 PMCID: PMC10327229 DOI: 10.1101/2023.06.23.23291824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
The MtrCDE efflux pump of Neisseria gonorrhoeae exports a wide range of antimicrobial compounds that the gonococcus encounters at mucosal surfaces during colonization and infection. Here, we evaluate the role of this efflux pump system in strain FA1090 in human male urethral infection with a Controlled Human Infection Model. Using the strategy of competitive multi-strain infection with wild-type FA1090 and an isogenic mutant strain that does not contain a functional MtrCDE pump, we found that the presence of the efflux pump during human experimental infection did not confer a competitive advantage. This finding is in contrast to previous findings in female mice, which demonstrated that gonococci of strain FA19 lacking a functional MtrCDE pump had a significantly reduced fitness compared to the wild type strain in the lower genital tract of female mice. We conducted competitive infections in female mice with FA19 and FA1090 strains, including mutants that do not assemble a functional Mtr efflux pump, demonstrating the fitness advantage provided byt the MtrCDE efflux pump during infection of mice is strain dependent. Our data indicate that new gonorrhea treatment strategies targeting the MtrCDE efflux pump functions may not be universally efficacious in naturally occurring infections. Owing to the equal fitness of FA1090 strains in men, our experiments unexpectedly demonstrated the likely presence of an early colonization bottleneck of N. gonorrhoeae in the human male urethra. TRIAL REGISTRATION Clinicaltrials.gov NCT03840811 .
Collapse
|
31
|
Apte S, Bhutda S, Ghosh S, Sharma K, Barton TE, Dibyachintan S, Sahay O, Roy S, Sinha AR, Adicherla H, Rakshit J, Tang S, Datey A, Santra S, Joseph J, Sasidharan S, Hammerschmidt S, Chakravortty D, Oggioni MR, Santra MK, Neill DR, Banerjee A. An innate pathogen sensing strategy involving ubiquitination of bacterial surface proteins. SCIENCE ADVANCES 2023; 9:eade1851. [PMID: 36947610 PMCID: PMC10032600 DOI: 10.1126/sciadv.ade1851] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 02/17/2023] [Indexed: 06/18/2023]
Abstract
Sensing of pathogens by ubiquitination is a critical arm of cellular immunity. However, universal ubiquitination targets on microbes remain unidentified. Here, using in vitro, ex vivo, and in vivo studies, we identify the first protein-based ubiquitination substrates on phylogenetically diverse bacteria by unveiling a strategy that uses recognition of degron-like motifs. Such motifs form a new class of intra-cytosolic pathogen-associated molecular patterns (PAMPs). Their incorporation enabled recognition of nonubiquitin targets by host ubiquitin ligases. We find that SCFFBW7 E3 ligase, supported by the regulatory kinase, glycogen synthase kinase 3β, is crucial for effective pathogen detection and clearance. This provides a mechanistic explanation for enhanced risk of infections in patients with chronic lymphocytic leukemia bearing mutations in F-box and WD repeat domain containing 7 protein. We conclude that exploitation of this generic pathogen sensing strategy allows conservation of host resources and boosts antimicrobial immunity.
Collapse
Affiliation(s)
- Shruti Apte
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| | - Smita Bhutda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| | - Sourav Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| | - Kuldeep Sharma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| | - Thomas E. Barton
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, L69 7BE Liverpool, UK
| | - Soham Dibyachintan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| | - Osheen Sahay
- Cancer Biology and Epigenetics Laboratory, National Centre for Cell Science, Ganeshkhind Road, Pune 411007, Maharashtra, India
| | - Suvapriya Roy
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| | - Akash Raj Sinha
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| | - Harikrishna Adicherla
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Habsiguda, Hyderabad 500007 Telangana, India
| | - Jyotirmoy Rakshit
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| | - Shiying Tang
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Akshay Datey
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru 560012, Karnataka, India
| | - Shweta Santra
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| | - Jincy Joseph
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| | - Sreeja Sasidharan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute of Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, D-17487 Greifswald, Germany
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru 560012, Karnataka, India
| | - Marco R. Oggioni
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Manas Kumar Santra
- Cancer Biology and Epigenetics Laboratory, National Centre for Cell Science, Ganeshkhind Road, Pune 411007, Maharashtra, India
| | - Daniel R. Neill
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, L69 7BE Liverpool, UK
| | - Anirban Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| |
Collapse
|
32
|
Kruckow KL, Zhao K, Bowdish DME, Orihuela CJ. Acute organ injury and long-term sequelae of severe pneumococcal infections. Pneumonia (Nathan) 2023; 15:5. [PMID: 36870980 PMCID: PMC9985869 DOI: 10.1186/s41479-023-00110-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 01/31/2023] [Indexed: 03/06/2023] Open
Abstract
Streptococcus pneumoniae (Spn) is a major public health problem, as it is a main cause of otitis media, community-acquired pneumonia, bacteremia, sepsis, and meningitis. Acute episodes of pneumococcal disease have been demonstrated to cause organ damage with lingering negative consequences. Cytotoxic products released by the bacterium, biomechanical and physiological stress resulting from infection, and the corresponding inflammatory response together contribute to organ damage accrued during infection. The collective result of this damage can be acutely life-threatening, but among survivors, it also contributes to the long-lasting sequelae of pneumococcal disease. These include the development of new morbidities or exacerbation of pre-existing conditions such as COPD, heart disease, and neurological impairments. Currently, pneumonia is ranked as the 9th leading cause of death, but this estimate only considers short-term mortality and likely underestimates the true long-term impact of disease. Herein, we review the data that indicates damage incurred during acute pneumococcal infection can result in long-term sequelae which reduces quality of life and life expectancy among pneumococcal disease survivors.
Collapse
Affiliation(s)
- Katherine L Kruckow
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kevin Zhao
- McMaster Immunology Research Centre and the Firestone Institute for Respiratory Health, McMaster University, Hamilton, Canada
| | - Dawn M E Bowdish
- McMaster Immunology Research Centre and the Firestone Institute for Respiratory Health, McMaster University, Hamilton, Canada
| | - Carlos J Orihuela
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
33
|
Jacques LC, Green AE, Barton TE, Baltazar M, Aleksandrowicz J, Xu R, Trochu E, Kadioglu A, Neill DR. Influence of Streptococcus pneumoniae Within-Strain Population Diversity on Virulence and Pathogenesis. Microbiol Spectr 2023; 11:e0310322. [PMID: 36507681 PMCID: PMC9927508 DOI: 10.1128/spectrum.03103-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
The short generation time of many bacterial pathogens allows the accumulation of de novo mutations during routine culture procedures used for the preparation and propagation of bacterial stocks. Taking the major human pathogen Streptococcus pneumoniae as an example, we sought to determine the influence of standard laboratory handling of microbes on within-strain genetic diversity and explore how these changes influence virulence characteristics and experimental outcomes. A single culture of S. pneumoniae D39 grown overnight resulted in the enrichment of previously rare genotypes present in bacterial freezer stocks and the introduction of new variation to the bacterial population through the acquisition of mutations. A comparison of D39 stocks from different laboratories demonstrated how changes in bacterial population structure taking place during individual culture events can cumulatively lead to fixed, divergent change that profoundly alters virulence characteristics. The passage of D39 through mouse models of infection, a process used to standardize virulence, resulted in the enrichment of high-fitness genotypes that were originally rare (<2% frequency) in D39 culture collection stocks and the loss of previously dominant genotypes. In the most striking example, the selection of a <2%-frequency genotype carrying a mutation in sdhB, a gene thought to be essential for the establishment of lung infection, was associated with enhanced systemic virulence. Three separately passaged D39 cultures originating from the same frozen stocks showed considerable genetic divergence despite comparable virulence. IMPORTANCE Laboratory bacteriology involves the use of high-density cultures that we often assume to be clonal but that in reality are populations consisting of multiple genotypes at various abundances. We have demonstrated that the genetic structure of a single population of a widely used Streptococcus pneumoniae strain can be substantially altered by even short-term laboratory handling and culture and that, over time, this can lead to changes in virulence characteristics. Our findings suggest that caution should be applied when comparing data generated in different laboratories using the same strain but also when comparing data within laboratories over time. Given the dramatic reductions in the cost of next-generation sequencing technology in recent years, we advocate for the frequent sampling and sequencing of bacterial isolate collections.
Collapse
Affiliation(s)
- Laura C. Jacques
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Angharad E. Green
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Thomas E. Barton
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Murielle Baltazar
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Julia Aleksandrowicz
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Rong Xu
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Erwan Trochu
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Aras Kadioglu
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Daniel R. Neill
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
34
|
Fritsch VN, Linzner N, Busche T, Said N, Weise C, Kalinowski J, Wahl MC, Antelmann H. The MerR-family regulator NmlR is involved in the defense against oxidative stress in Streptococcus pneumoniae. Mol Microbiol 2023; 119:191-207. [PMID: 36349475 DOI: 10.1111/mmi.14999] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/25/2022] [Accepted: 10/30/2022] [Indexed: 11/11/2022]
Abstract
Streptococcus pneumoniae has to cope with the strong oxidant hypochlorous acid (HOCl), during host-pathogen interactions. Thus, we analyzed the global gene expression profile of S. pneumoniae D39 towards HOCl stress. In the RNA-seq transcriptome, the NmlR, SifR, CtsR, HrcA, SczA and CopY regulons and the etrx1-ccdA1-msrAB2 operon were most strongly induced under HOCl stress, which participate in the oxidative, electrophile and metal stress response in S. pneumoniae. The MerR-family regulator NmlR harbors a conserved Cys52 and controls the alcohol dehydrogenase-encoding adhC gene under carbonyl and NO stress. We demonstrated that NmlR senses also HOCl stress to activate transcription of the nmlR-adhC operon. HOCl-induced transcription of adhC required Cys52 of NmlR in vivo. Using mass spectrometry, NmlR was shown to be oxidized to intersubunit disulfides or S-glutathionylated under oxidative stress in vitro. A broccoli-FLAP-based assay further showed that both NmlR disulfides significantly increased transcription initiation at the nmlR promoter by RNAP in vitro, which depends on Cys52. Phenotype analyses revealed that NmlR functions in the defense against oxidative stress and promotes survival of S. pneumoniae during macrophage infections. In conclusion, NmlR was characterized as HOCl-sensing transcriptional regulator, which activates transcription of adhC under oxidative stress by thiol switches in S. pneumoniae.
Collapse
Affiliation(s)
| | - Nico Linzner
- Institute of Biology-Microbiology, Freie Universität Berlin, Berlin, Germany
| | - Tobias Busche
- Center for Biotechnology, University Bielefeld, Bielefeld, Germany.,NGS Core Facility, Medical School OWL, Bielefeld University, Bielefeld, Germany
| | - Nelly Said
- Laboratory of Structural Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Christoph Weise
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Jörn Kalinowski
- Center for Biotechnology, University Bielefeld, Bielefeld, Germany
| | - Markus C Wahl
- Laboratory of Structural Biochemistry, Freie Universität Berlin, Berlin, Germany.,Macromolecular Crystallography, Helmholtz-Zentrum Berlin für Materialien und Energie, Berlin, Germany
| | - Haike Antelmann
- Institute of Biology-Microbiology, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
35
|
Feng W, Chittò M, Moriarty TF, Li G, Wang X. Targeted Drug Delivery Systems for Eliminating Intracellular Bacteria. Macromol Biosci 2023; 23:e2200311. [PMID: 36189899 DOI: 10.1002/mabi.202200311] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/08/2022] [Indexed: 01/19/2023]
Abstract
The intracellular survival of pathogenic bacteria requires a range of survival strategies and virulence factors. These infections are a significant clinical challenge, wherein treatment frequently fails because of poor antibiotic penetration, stability, and retention in host cells. Drug delivery systems (DDSs) are promising tools to overcome these shortcomings and enhance the efficacy of antibiotic therapy. In this review, the classification and the mechanisms of intracellular bacterial persistence are elaborated. Furthermore, the systematic design strategies applied to DDSs to eliminate intracellular bacteria are also described, and the strategies used for internalization, intracellular activation, bacterial targeting, and immune enhancement are highlighted. Finally, this overview provides guidance for constructing functionalized DDSs to effectively eliminate intracellular bacteria.
Collapse
Affiliation(s)
- Wenli Feng
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China.,AO Research Institute Davos, Davos, 7270, Switzerland
| | - Marco Chittò
- AO Research Institute Davos, Davos, 7270, Switzerland
| | | | - Guofeng Li
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Xing Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| |
Collapse
|
36
|
Hernandez-Morfa M, Reinoso-Vizcaíno NM, Olivero NB, Zappia VE, Cortes PR, Jaime A, Echenique J. Host Cell Oxidative Stress Promotes Intracellular Fluoroquinolone Persisters of Streptococcus pneumoniae. Microbiol Spectr 2022; 10:e0436422. [PMID: 36445159 PMCID: PMC9769771 DOI: 10.1128/spectrum.04364-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 12/02/2022] Open
Abstract
Bacterial persisters represent a small subpopulation that tolerates high antibiotic concentrations without acquiring heritable resistance, and it may be generated by environmental factors. Here, we report the first antibiotic persistence mechanism in Streptococcus pneumoniae, which is induced by oxidative stress conditions and allows the pneumococcus to survive in the presence of fluoroquinolones. We demonstrated that fluoroquinolone persistence is prompted by both the impact of growth arrest and the oxidative stress response induced by H2O2 in bacterial cells. This process protected pneumococci against the deleterious effects of high ROS levels induced by fluoroquinolones. Importantly, S. pneumoniae develops persistence during infection, and is dependent on the oxidative stress status of the host cells, indicating that its transient intracellular life contributes to this mechanism. Furthermore, our findings suggest persistence may influence the outcome of antibiotic therapy and be part of a multistep mechanism in the evolution of fluoroquinolone resistance. IMPORTANCE In S. pneumoniae, different mechanisms that counteract antibiotic effects have been described, such as vancomycin tolerance, heteroresistance to penicillin and fluoroquinolone resistance, which critically affect the therapeutic efficacy. Antibiotic persistence is a type of antibiotic tolerance that allows a bacterial subpopulation to survive lethal antimicrobial concentrations. In this work, we used a host-cell infection model to reveal fluoroquinolone persistence in S. pneumoniae. This mechanism is induced by oxidative stress that the pneumococcus must overcome to survive in host cells. Many fluoroquinolones, such as levofloxacin and moxifloxacin, have a broad spectrum of activity against bacterial pathogens of community-acquired pneumonia, and they are used to treat pneumococcal diseases. However, the emergence of fluoroquinolone-resistant strains complicates antibiotic treatment of invasive infections. Consequently, antibiotic persistence in S. pneumoniae is clinically relevant due to prolonged exposure to fluoroquinolones likely favors the acquisition of mutations that generate antibiotic resistance in persisters. In addition, this work contributes to the knowledge of antibiotic persistence mechanisms in bacteria.
Collapse
Affiliation(s)
- Mirelys Hernandez-Morfa
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Nicolás M. Reinoso-Vizcaíno
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Nadia B. Olivero
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Victoria E. Zappia
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Paulo R. Cortes
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Andrea Jaime
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - José Echenique
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
37
|
Streptococcus agalactiae npx Is Required for Survival in Human Placental Macrophages and Full Virulence in a Model of Ascending Vaginal Infection during Pregnancy. mBio 2022; 13:e0287022. [PMID: 36409087 PMCID: PMC9765263 DOI: 10.1128/mbio.02870-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Streptococcus agalactiae, also known as group B Streptococcus (GBS), is a Gram-positive encapsulated bacterium that colonizes the gastrointestinal tract of 30 to 50% of humans. GBS causes invasive infection during pregnancy that can lead to chorioamnionitis, funisitis, preterm prelabor rupture of membranes (PPROM), preterm birth, neonatal sepsis, and maternal and fetal demise. Upon infecting the host, GBS encounters sentinel innate immune cells, such as macrophages, within reproductive tissues. Once phagocytosed by macrophages, GBS upregulates the expression of the gene npx, which encodes an NADH peroxidase. GBS mutants with an npx deletion (Δnpx) are exquisitely sensitive to reactive oxygen stress. Furthermore, we have shown that npx is required for GBS survival in both THP-1 and placental macrophages. In an in vivo murine model of ascending GBS vaginal infection during pregnancy, npx is required for invading reproductive tissues and is critical for inducing disease progression, including PPROM and preterm birth. Reproductive tissue cytokine production was also significantly diminished in Δnpx mutant-infected animals compared to that in animals infected with wild-type (WT) GBS. Complementation in trans reversed this phenotype, indicating that npx is critical for GBS survival and the initiation of proinflammatory signaling in the gravid host. IMPORTANCE This study sheds new light on the way that group B Streptococcus (GBS) defends itself against oxidative stress in the infected host. The enzyme encoded by the GBS gene npx is an NADH peroxidase that, our study reveals, provides defense against macrophage-derived reactive oxygen stress and facilitates infections of the uterus during pregnancy. This enzyme could represent a tractable target for future treatment strategies against invasive GBS infections.
Collapse
|
38
|
Yesilkaya H, Oggioni MR, Andrew PW. Streptococcus pneumoniae: 'captain of the men of death' and financial burden. MICROBIOLOGY (READING, ENGLAND) 2022; 168. [PMID: 36748691 DOI: 10.1099/mic.0.001275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Streptococcus pneumoniae may inhabit the upper respiratory tract of humans without causing harm but it also causes diseases with high morbidity and mortality. It has excellent adaptive capabilities thanks to its ability to shuffle its genetic content by acquiring and incorporating DNA from other bacteria and is highly competent for genetic transformation. Sugar sensing, cleavage and transport ensure its fitness and survival in the host, and intracellular survival in macrophages has been linked to virulence. The polysaccharide capsule and toxin pneumolysin are the most important virulence determinants. Polysaccharide-based vaccines provide protection against the serotypes represented in vaccine formulations.
Collapse
Affiliation(s)
- Hasan Yesilkaya
- Department of Respiratory Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Marco R Oggioni
- Department of Genetics, University of Leicester, Leicester LE1 7RH, UK.,Dipartimento di Farmacia e Biotecnologie, University of Bologna, Bologna, Italy
| | - Peter W Andrew
- Department of Respiratory Sciences, University of Leicester, Leicester LE1 7RH, UK
| |
Collapse
|
39
|
Aleith J, Brendel M, Weipert E, Müller M, Schultz D, Müller-Hilke B. Influenza A Virus Exacerbates Group A Streptococcus Infection and Thwarts Anti-Bacterial Inflammatory Responses in Murine Macrophages. Pathogens 2022; 11:1320. [PMID: 36365071 PMCID: PMC9699311 DOI: 10.3390/pathogens11111320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 09/30/2023] Open
Abstract
Seasonal influenza epidemics pose a considerable hazard for global health. In the past decades, accumulating evidence revealed that influenza A virus (IAV) renders the host vulnerable to bacterial superinfections which in turn are a major cause for morbidity and mortality. However, whether the impact of influenza on anti-bacterial innate immunity is restricted to the vicinity of the lung or systemically extends to remote sites is underexplored. We therefore sought to investigate intranasal infection of adult C57BL/6J mice with IAV H1N1 in combination with bacteremia elicited by intravenous application of Group A Streptococcus (GAS). Co-infection in vivo was supplemented in vitro by challenging murine bone marrow derived macrophages and exploring gene expression and cytokine secretion. Our results show that viral infection of mice caused mild disease and induced the depletion of CCL2 in the periphery. Influenza preceding GAS infection promoted the occurrence of paw edemas and was accompanied by exacerbated disease scores. In vitro co-infection of macrophages led to significantly elevated expression of TLR2 and CD80 compared to bacterial mono-infection, whereas CD163 and CD206 were downregulated. The GAS-inducible upregulation of inflammatory genes, such as Nos2, as well as the secretion of TNFα and IL-1β were notably reduced or even abrogated following co-infection. Our results indicate that IAV primes an innate immune layout that is inadequately equipped for bacterial clearance.
Collapse
Affiliation(s)
- Johann Aleith
- Core Facility for Cell Sorting and Cell Analysis, Rostock University Medical Center, 18057 Rostock, Germany
| | - Maria Brendel
- Core Facility for Cell Sorting and Cell Analysis, Rostock University Medical Center, 18057 Rostock, Germany
| | - Erik Weipert
- Core Facility for Cell Sorting and Cell Analysis, Rostock University Medical Center, 18057 Rostock, Germany
| | - Michael Müller
- Core Facility for Cell Sorting and Cell Analysis, Rostock University Medical Center, 18057 Rostock, Germany
| | - Daniel Schultz
- Institute of Biochemistry, University of Greifswald, 17489 Greifswald, Germany
| | - Ko-Infekt Study Group
- Institute of Biochemistry, University of Greifswald, 17489 Greifswald, Germany
- Institute of Immunology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
- Institute of Medical Microbiology, Virology and Hygiene, Rostock University Medical Center, 18057 Rostock, Germany
| | - Brigitte Müller-Hilke
- Core Facility for Cell Sorting and Cell Analysis, Rostock University Medical Center, 18057 Rostock, Germany
| |
Collapse
|
40
|
Zhang Y, Liu X, Klionsky DJ, Lu B, Zhong Q. Manipulating autophagic degradation in human diseases: from mechanisms to interventions. LIFE MEDICINE 2022; 1:120-148. [PMID: 39871921 PMCID: PMC11749641 DOI: 10.1093/lifemedi/lnac043] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/08/2022] [Indexed: 01/29/2025]
Abstract
Targeted degradation, having emerged as a powerful and promising strategy in drug discovery in the past two decades, has provided a solution for many once undruggable targets involved in various diseases. While earlier targeted degradation tools, as exemplified by PROteolysis-TArgeting Chimera (PROTAC), focused on harnessing the ubiquitin-proteasome system, novel approaches that aim to utilize autophagy, a potent, lysosome-dependent degradation pathway, have also surfaced recently as promising modalities. In this review, we first introduce the mechanisms that establish selectivity in autophagy, which provides the rationales for autophagy-based targeted degradation; we also provide an overview on the panoply of cellular machinery involved in this process, an arsenal that could be potentially harnessed. On this basis, we propose four strategies for designing autophagy-based targeted degraders, including Tagging Targets, Directly Engaging Targets, Initiating Autophagy at Targets, and Phagophore-Tethering to Targets. We introduce the current frontiers in this field, including AUtophagy-TArgeting Chimera (AUTAC), Targeted Protein Autophagy (TPA), AUTOphagy-TArgeting Chimera (AUTOTAC, not to be confused with AUTAC), AuTophagosome TEthering Compound (ATTEC), and other experimental approaches as case studies for each strategy. Finally, we put forward a workflow for generating autophagy-based degraders and some important questions that may guide and inspire the process.
Collapse
Affiliation(s)
- Yiqing Zhang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 20025, China
| | - Xiaoxia Liu
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 20025, China
| | - Daniel J Klionsky
- Department of Molecular, Cellular, and Developmental Biology, and the Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109-2216, USA
| | - Boxun Lu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Huashan Hospital, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Qing Zhong
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 20025, China
| |
Collapse
|
41
|
Zlotnikov ID, Ezhov AA, Petrov RA, Vigovskiy MA, Grigorieva OA, Belogurova NG, Kudryashova EV. Mannosylated Polymeric Ligands for Targeted Delivery of Antibacterials and Their Adjuvants to Macrophages for the Enhancement of the Drug Efficiency. Pharmaceuticals (Basel) 2022; 15:1172. [PMID: 36297284 PMCID: PMC9607288 DOI: 10.3390/ph15101172] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/16/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Bacterial infections and especially resistant strains of pathogens localized in macrophages and granulomas are intractable diseases that pose a threat to millions of people. In this paper, the theoretical and experimental foundations for solving this problem are proposed due to two key aspects. The first is the use of a three-component polymer system for delivering fluoroquinolones to macrophages due to high-affinity interaction with mannose receptors (CD206). Cytometry assay determined that 95.5% macrophage-like cells were FITC-positive after adding high-affine to CD206 trimannoside conjugate HPCD-PEI1.8-triMan, and 61.7% were FITC-positive after adding medium-affine ligand with linear mannose label HPCD-PEI1.8-Man. The second aspect is the use of adjuvants, which are synergists for antibiotics. Using FTIR and NMR spectroscopy, it was shown that molecular containers, namely mannosylated polyethyleneimines (PEIs) and cyclodextrins (CDs), load moxifloxacin (MF) with dissociation constants of the order of 10-4-10-6 M; moreover, due to prolonged release and adsorption on the cell membrane, they enhance the effect of MF. Using CLSM, it was shown that eugenol (EG) increases the penetration of doxorubicin (Dox) into cells by an order of magnitude due to the creation of defects in the bacterial wall and the inhibition of efflux proteins. Fluorescence spectroscopy showed that 0.5% EG penetrates into bacteria and inhibits efflux proteins, which makes it possible to increase the maximum concentration of the antibiotic by 60% and maintain it for several hours until the pathogens are completely neutralized. Regulation of efflux is a possible way to overcome multiple drug resistance of both pathogens and cancer cells.
Collapse
Affiliation(s)
- Igor D. Zlotnikov
- Faculty of Chemistry, Lomonosov Moscow State University, Leninskie Gory, 1/3, 119991 Moscow, Russia
| | - Alexander A. Ezhov
- Faculty of Physics, Lomonosov Moscow State University, Leninskie Gory, 1/2, 119991 Moscow, Russia
| | - Rostislav A. Petrov
- Faculty of Chemistry, Lomonosov Moscow State University, Leninskie Gory, 1/3, 119991 Moscow, Russia
| | - Maksim A. Vigovskiy
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 27/10, Lomonosovsky Ave., 119192 Moscow, Russia
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia
| | - Olga A. Grigorieva
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 27/10, Lomonosovsky Ave., 119192 Moscow, Russia
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia
| | - Natalya G. Belogurova
- Faculty of Chemistry, Lomonosov Moscow State University, Leninskie Gory, 1/3, 119991 Moscow, Russia
| | - Elena V. Kudryashova
- Faculty of Chemistry, Lomonosov Moscow State University, Leninskie Gory, 1/3, 119991 Moscow, Russia
| |
Collapse
|
42
|
Nyazika TK, Sibale L, Phiri J, De Ste Croix M, Jasiunaite Z, Mkandawire C, Malamba R, Kankwatira A, Manduwa M, Ferreira DM, Nyirenda TS, Oggioni MR, Mwandumba HC, Jambo KC. Intracellular survival of Streptococcus pneumoniae in human alveolar macrophages is augmented with HIV infection. Front Immunol 2022; 13:992659. [PMID: 36203580 PMCID: PMC9531125 DOI: 10.3389/fimmu.2022.992659] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/31/2022] [Indexed: 11/22/2022] Open
Abstract
People Living with HIV (PLHIV) are at an increased risk of pneumococcal pneumonia than HIV-uninfected adults, but the reasons for this are still not well understood. We investigated whether alveolar macrophages (AM) mediated control of pneumococcal infection is impaired in PLHIV compared to HIV-uninfected adults. We assessed anti-bactericidal activity against Streptococcus pneumoniae of primary human AM obtained from PLHIV and HIV-uninfected adults. We found that pneumococcus survived intracellularly in AMs at least 24 hours post ex vivo infection, and this was more frequent in PLHIV than HIV-uninfected adults. Corroborating these findings, in vivo evidence showed that PLHIV had a higher propensity for harboring S. pneumoniae within their AMs than HIV-uninfected adults. Moreover, bacterial intracellular survival in AMs was associated with extracellular propagation of pneumococcal infection. Our data suggest that failure of AMs to eliminate S. pneumoniae intracellularly could contribute to the increased risk of pneumococcal pneumonia in PLHIV.
Collapse
Affiliation(s)
- Tinashe K. Nyazika
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Lusako Sibale
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi
| | - Joseph Phiri
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi
| | - Megan De Ste Croix
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Zydrune Jasiunaite
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Christopher Mkandawire
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi
| | - Rose Malamba
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi
| | - Anstead Kankwatira
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi
| | - Miriam Manduwa
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi
| | - Daniela M. Ferreira
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Tonney S. Nyirenda
- Department of Pathology, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Marco R. Oggioni
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
- Dipartimento di Farmacia e Biotecnologie, Universita di Bologna, Bologna, Italy
| | - Henry C. Mwandumba
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Kondwani C. Jambo
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, University of Malawi College of Medicine, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| |
Collapse
|
43
|
Korir ML, Doster RS, Lu J, Guevara MA, Spicer SK, Moore RE, Francis JD, Rogers LM, Haley KP, Blackman A, Noble KN, Eastman AJ, Williams JA, Damo SM, Boyd KL, Townsend SD, Henrique Serezani C, Aronoff DM, Manning SD, Gaddy JA. Streptococcus agalactiae cadD alleviates metal stress and promotes intracellular survival in macrophages and ascending infection during pregnancy. Nat Commun 2022; 13:5392. [PMID: 36104331 PMCID: PMC9474517 DOI: 10.1038/s41467-022-32916-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 08/24/2022] [Indexed: 01/17/2023] Open
Abstract
Perinatal infection with Streptococcus agalactiae, or Group B Streptococcus (GBS), is associated with preterm birth, neonatal sepsis, and stillbirth. Here, we study the interactions of GBS with macrophages, essential sentinel immune cells that defend the gravid reproductive tract. Transcriptional analyses of GBS-macrophage co-cultures reveal enhanced expression of a gene encoding a putative metal resistance determinant, cadD. Deletion of cadD reduces GBS survival in macrophages, metal efflux, and resistance to metal toxicity. In a mouse model of ascending infection during pregnancy, the ΔcadD strain displays attenuated bacterial burden, inflammation, and cytokine production in gestational tissues. Furthermore, depletion of host macrophages alters cytokine expression and decreases GBS invasion in a cadD-dependent fashion. Our results indicate that GBS cadD plays an important role in metal detoxification, which promotes immune evasion and bacterial proliferation in the pregnant host.
Collapse
Affiliation(s)
- Michelle L Korir
- Michigan State University, Department of Microbiology and Molecular Genetics, East Lansing, MI, USA
- Aurora University, Department of Biology, Aurora, IL, USA
| | - Ryan S Doster
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Jacky Lu
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Stanford University, Palo Alto, CA, USA
| | - Miriam A Guevara
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sabrina K Spicer
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Rebecca E Moore
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Jamisha D Francis
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lisa M Rogers
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kathryn P Haley
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Sciences, Grand Valley State University, Allendale, MI, USA
| | - Amondrea Blackman
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kristen N Noble
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alison J Eastman
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Janice A Williams
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Steven M Damo
- Department of Life and Physical Sciences, Fisk University, Nashville, TN, USA
- Department of Biochemistry and Structural Biology, Vanderbilt University, Nashville, TN, USA
| | - Kelli L Boyd
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - C Henrique Serezani
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - David M Aronoff
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shannon D Manning
- Michigan State University, Department of Microbiology and Molecular Genetics, East Lansing, MI, USA.
| | - Jennifer A Gaddy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Center for Medicine, Health, and Society, Vanderbilt University, Nashville, TN, USA.
- Department of Veterans Affairs, Tennessee Valley Healthcare Systems, Nashville, TN, USA.
| |
Collapse
|
44
|
Song L, Hu X, Ren X, Liu J, Liu X. Antibacterial Modes of Herbal Flavonoids Combat Resistant Bacteria. Front Pharmacol 2022; 13:873374. [PMID: 35847042 PMCID: PMC9278433 DOI: 10.3389/fphar.2022.873374] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/17/2022] [Indexed: 12/21/2022] Open
Abstract
The increasing dissemination of multidrug resistant (MDR) bacterial infections endangers global public health. How to develop effective antibacterial agents against resistant bacteria is becoming one of the most urgent demands to solve the drug resistance crisis. Traditional Chinese medicine (TCM) with multi-target antibacterial actions are emerging as an effective way to combat the antibacterial resistance. Based on the innovative concept of organic wholeness and syndrome differentiation, TCM use in antibacterial therapies is encouraging. Herein, advances on flavonoid compounds of heat-clearing Chinese medicine exhibit their potential for the therapy of resistant bacteria. In this review, we focus on the antibacterial modes of herbal flavonoids. Additionally, we overview the targets of flavonoid compounds and divide them into direct-acting antibacterial compounds (DACs) and host-acting antibacterial compounds (HACs) based on their modes of action. We also discuss the associated functional groups of flavonoid compounds and highlight recent pharmacological activities against diverse resistant bacteria to provide the candidate drugs for the clinical infection.
Collapse
Affiliation(s)
- Lianyu Song
- Beijing Traditional Chinese Veterinary Engineering Center and Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Changping, China
| | - Xin Hu
- Animal Science and Technology College, Beijing University of Agriculture, Changping, China
| | - Xiaomin Ren
- Beijing Traditional Chinese Veterinary Engineering Center and Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Changping, China
| | - Jing Liu
- Animal Science and Technology College, Beijing University of Agriculture, Changping, China
| | - Xiaoye Liu
- Beijing Traditional Chinese Veterinary Engineering Center and Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Changping, China
- Animal Science and Technology College, Beijing University of Agriculture, Changping, China
- *Correspondence: Xiaoye Liu,
| |
Collapse
|
45
|
Hames RG, Jasiunaite Z, Ercoli G, Wanford JJ, Carreno D, Straatman K, Martinez-Pomares L, Yesilkaya H, Glenn S, Moxon ER, Andrew PW, Kyriacou CP, Oggioni MR. Diurnal Differences in Intracellular Replication Within Splenic Macrophages Correlates With the Outcome of Pneumococcal Infection. Front Immunol 2022; 13:907461. [PMID: 35720383 PMCID: PMC9201068 DOI: 10.3389/fimmu.2022.907461] [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: 03/29/2022] [Accepted: 05/09/2022] [Indexed: 12/04/2022] Open
Abstract
Circadian rhythms affect the progression and severity of bacterial infections including those caused by Streptococcus pneumoniae, but the mechanisms responsible for this phenomenon remain largely elusive. Following advances in our understanding of the role of replication of S. pneumoniae within splenic macrophages, we sought to investigate whether events within the spleen correlate with differential outcomes of invasive pneumococcal infection. Utilising murine invasive pneumococcal disease (IPD) models, here we report that infection during the murine active phase (zeitgeber time 15; 15h after start of light cycle, 3h after start of dark cycle) resulted in significantly faster onset of septicaemia compared to rest phase (zeitgeber time 3; 3h after start of light cycle) infection. This correlated with significantly higher pneumococcal burden within the spleen of active phase-infected mice at early time points compared to rest phase-infected mice. Whole-section confocal microscopy analysis of these spleens revealed that the number of pneumococci is significantly higher exclusively within marginal zone metallophilic macrophages (MMMs) known to allow intracellular pneumococcal replication as a prerequisite step to the onset of septicaemia. Pneumococcal clusters within MMMs were more abundant and increased in size over time in active phase-infected mice compared to those in rest phase-infected mice which decreased in size and were present in a lower percentage of MMMs. This phenomenon preceded significantly higher levels of bacteraemia alongside serum IL-6 and TNF-α concentrations in active phase-infected mice following re-seeding of pneumococci into the blood. These data greatly advance our fundamental knowledge of pneumococcal infection by linking susceptibility to invasive pneumococcal infection to variation in the propensity of MMMs to allow persistence and replication of phagocytosed bacteria. These findings also outline a somewhat rare scenario whereby the active phase of an organism’s circadian cycle plays a seemingly counterproductive role in the control of invasive infection.
Collapse
Affiliation(s)
- Ryan G Hames
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Zydrune Jasiunaite
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Giuseppe Ercoli
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Division of Medicine, University College Medical School, London, United Kingdom
| | - Joseph J Wanford
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - David Carreno
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Kornelis Straatman
- Advanced Imaging Facility, University of Leicester, Leicester, United Kingdom
| | | | - Hasan Yesilkaya
- Department of Respiratory Sciences, University of Leicester, Leicester, United Kingdom
| | - Sarah Glenn
- Preclinical Research Facility, University of Leicester, Leicester, United Kingdom
| | - E Richard Moxon
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Peter W Andrew
- Department of Respiratory Sciences, University of Leicester, Leicester, United Kingdom
| | - Charalambos P Kyriacou
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Marco R Oggioni
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom.,Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| |
Collapse
|
46
|
Moscardini IF, Santoro F, Carraro M, Gerlini A, Fiorino F, Germoni C, Gholami S, Pettini E, Medaglini D, Iannelli F, Pozzi G. Immune Memory After Respiratory Infection With Streptococcus pneumoniae Is Revealed by in vitro Stimulation of Murine Splenocytes With Inactivated Pneumococcal Whole Cells: Evidence of Early Recall Responses by Transcriptomic Analysis. Front Cell Infect Microbiol 2022; 12:869763. [PMID: 35795182 PMCID: PMC9251119 DOI: 10.3389/fcimb.2022.869763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 04/21/2022] [Indexed: 11/13/2022] Open
Abstract
The in vitro stimulation of immune system cells with live or killed bacteria is essential for understanding the host response to pathogens. In the present study, we propose a model combining transcriptomic and cytokine assays on murine splenocytes to describe the immune recall in the days following pneumococcal lung infection. Mice were sacrificed at days 1, 2, 4, and 7 after Streptococcus pneumoniae (TIGR4 serotype 4) intranasal infection and splenocytes were cultured in the presence or absence of the same inactivated bacterial strain to access the transcriptomic and cytokine profiles. The stimulation of splenocytes from infected mice led to a higher number of differentially expressed genes than the infection or stimulation alone, resulting in the enrichment of 40 unique blood transcription modules, including many pathways related to adaptive immunity and cytokines. Together with transcriptomic data, cytokines levels suggested the presence of a recall immune response promoting both innate and adaptive immunity, stronger from the fourth day after infection. Dimensionality reduction and feature selection identified key variables of this recall response and the genes associated with the increase in cytokine concentrations. This model could study the immune responses involved in pneumococcal infection and possibly monitor vaccine immune response and experimental therapies efficacy in future studies.
Collapse
Affiliation(s)
| | - Francesco Santoro
- Laboratory of Molecular Microbiology and Biotechnology (LAMMB), Department of Medical Biotechnologies, University of Siena, Siena, Italy
- *Correspondence: Francesco Santoro,
| | - Monica Carraro
- Laboratory of Molecular Microbiology and Biotechnology (LAMMB), Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | | | - Fabio Fiorino
- Laboratory of Molecular Microbiology and Biotechnology (LAMMB), Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Chiara Germoni
- Laboratory of Molecular Microbiology and Biotechnology (LAMMB), Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Samaneh Gholami
- Laboratory of Molecular Microbiology and Biotechnology (LAMMB), Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Elena Pettini
- Laboratory of Molecular Microbiology and Biotechnology (LAMMB), Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology (LAMMB), Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Francesco Iannelli
- Laboratory of Molecular Microbiology and Biotechnology (LAMMB), Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Gianni Pozzi
- Laboratory of Molecular Microbiology and Biotechnology (LAMMB), Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
47
|
Phagosomal Acidification Is Required to Kill Streptococcus pneumoniae in a Zebrafish Model. Cell Microbiol 2022. [DOI: 10.1155/2022/9429516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Streptococcus pneumoniae (the pneumococcus) is a major human pathogen causing invasive disease, including community-acquired bacteraemia, and remains a leading cause of global mortality. Understanding the role of phagocytes in killing bacteria is still limited, especially in vivo. In this study, we established a zebrafish model to study the interaction between intravenously administered pneumococci and professional phagocytes such as macrophages and neutrophils, to unravel bacterial killing mechanisms employed by these immune cells. Our model confirmed the key role of polysaccharide capsule in promoting pneumococcal virulence through inhibition of phagocytosis. Conversely, we show pneumococci lacking a capsule are rapidly internalised by macrophages. Low doses of encapsulated S. pneumoniae cause near 100% mortality within 48 hours postinfection (hpi), while 50 times higher doses of unencapsulated pneumococci are easily cleared. Time course analysis of in vivo bacterial numbers reveals that while encapsulated pneumococcus proliferates to levels exceeding 105 CFU at the time of host death, unencapsulated bacteria are unable to grow and are cleared within 20 hpi. Using genetically induced macrophage depletion, we confirmed an essential role for macrophages in bacterial clearance. Additionally, we show that upon phagocytosis by macrophages, phagosomes undergo rapid acidification. Genetic and chemical inhibition of vacuolar ATPase (v-ATPase) prevents intracellular bacterial killing and induces host death indicating a key role of phagosomal acidification in immunity to invading pneumococci. We also show that our model can be used to study the efficacy of antimicrobials against pneumococci in vivo. Collectively, our data confirm that larval zebrafish can be used to dissect killing mechanisms during pneumococcal infection in vivo and highlight key roles for phagosomal acidification in macrophages for pathogen clearance.
Collapse
|
48
|
Klimova N, Holubova J, Streparola G, Tomala J, Brazdilova L, Stanek O, Bumba L, Sebo P. Pertussis toxin suppresses dendritic cell-mediated delivery of B. pertussis into lung-draining lymph nodes. PLoS Pathog 2022; 18:e1010577. [PMID: 35666769 PMCID: PMC9216613 DOI: 10.1371/journal.ppat.1010577] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/22/2022] [Accepted: 05/08/2022] [Indexed: 11/18/2022] Open
Abstract
The adenylate cyclase (ACT) and the pertussis (PT) toxins of Bordetella pertussis exert potent immunomodulatory activities that synergize to suppress host defense in the course of whooping cough pathogenesis. We compared the mouse lung infection capacities of B. pertussis (Bp) mutants (Bp AC− or Bp PT–) producing enzymatically inactive toxoids and confirm that ACT action is required for maximal bacterial proliferation in the first days of infection, whereas PT action is crucial for persistence of B. pertussis in mouse lungs. Despite accelerated and near complete clearance from the lungs by day 14 of infection, the PT− bacteria accumulated within the lymphoid tissue of lung-draining mediastinal lymph nodes (mLNs). In contrast, the wild type or AC− bacteria colonized the lungs but did not enter into mLNs. Lung infection by the PT− mutant triggered an early arrival of migratory conventional dendritic cells with associated bacteria into mLNs, where the PT− bacteria entered the T cell-rich paracortex of mLNs by day 5 and proliferated in clusters within the B-cell zone (cortex) of mLNs by day 14, being eventually phagocytosed by infiltrating neutrophils. Finally, only infection by the PT− bacteria triggered an early production of anti-B. pertussis serum IgG antibodies already within 14 days of infection. These results reveal that action of the pertussis toxin blocks DC-mediated delivery of B. pertussis bacteria into mLNs and prevents bacterial colonization of mLNs, thus hampering early adaptive immune response to B. pertussis infection. Of the three classical Bordetella species causing respiratory infections in mammals, only the human-specialized whooping cough agent B. pertussis produces the pertussis toxin (PT) as its major virulence factor. Human pertussis is an acute respiratory illness and the pleiotropic activities of pertussis toxin account for the characteristic systemic manifestations of the disease, such as hyperleukocytosis, histamine sensitization, hyperinsulinemia, or inflammatory lung pathology. We found that PT activity inhibits the migration of infected dendritic cells from the lungs into the draining mediastinal lymph nodes (mLNs). This prevents mLN infection by bacteria evading from migratory cells and delivery of bacterial antigens into mLNs. As a result, the induction of adaptive serum antibody responses to infection is delayed. We thus propose that PT action serves to create a time window for proliferation of B. pertussis on airway mucosa to facilitate transmission of the pathogen among humans.
Collapse
Affiliation(s)
- Nela Klimova
- Institute of Microbiology of the Czech Academy of Sciences,Prague, Czech Republic
- Faculty of Sciences, Charles University, Prague, Czech Republic
| | - Jana Holubova
- Institute of Microbiology of the Czech Academy of Sciences,Prague, Czech Republic
| | - Gaia Streparola
- Institute of Microbiology of the Czech Academy of Sciences,Prague, Czech Republic
- Czech Centre for Phenogenomics BIOCEV, Vestec, Czech Republic
| | - Jakub Tomala
- Institute of Microbiology of the Czech Academy of Sciences,Prague, Czech Republic
- Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Ludmila Brazdilova
- Institute of Microbiology of the Czech Academy of Sciences,Prague, Czech Republic
- Faculty of Sciences, Charles University, Prague, Czech Republic
| | - Ondrej Stanek
- Institute of Microbiology of the Czech Academy of Sciences,Prague, Czech Republic
| | - Ladislav Bumba
- Institute of Microbiology of the Czech Academy of Sciences,Prague, Czech Republic
- * E-mail: (LB); (PS)
| | - Peter Sebo
- Institute of Microbiology of the Czech Academy of Sciences,Prague, Czech Republic
- * E-mail: (LB); (PS)
| |
Collapse
|
49
|
Pneumococcal Phasevarions Control Multiple Virulence Traits, Including Vaccine Candidate Expression. Microbiol Spectr 2022; 10:e0091622. [PMID: 35536022 PMCID: PMC9241608 DOI: 10.1128/spectrum.00916-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Streptococcus pneumoniae is the most common cause of bacterial illness worldwide. Current vaccines based on the polysaccharide capsule are only effective against a limited number of the >100 capsular serotypes. A universal vaccine based on conserved protein antigens requires a thorough understanding of gene expression in S. pneumoniae. All S. pneumoniae strains encode the SpnIII Restriction-Modification system. This system contains a phase-variable methyltransferase that switches specificity, and controls expression of multiple genes—a phasevarion. We examined the role of this phasevarion during pneumococcal pathobiology, and determined if phase variation resulted in differences in expression of currently investigated conserved protein antigens. Using locked strains that express a single methyltransferase specificity, we found differences in clinically relevant traits, including survival in blood, and adherence to and invasion of human cells. We also observed differences in expression of numerous proteinaceous vaccine candidates, which complicates selection of antigens for inclusion in a universal protein-based pneumococcal vaccine. This study will inform vaccine design against S. pneumoniae by ensuring only stably expressed candidates are included in a rationally designed vaccine. IMPORTANCES. pneumoniae is the world’s foremost bacterial pathogen. S. pneumoniae encodes a phasevarion (phase-variable regulon), that results in differential expression of multiple genes. Previous work demonstrated that the pneumococcal SpnIII phasevarion switches between six different expression states, generating six unique phenotypic variants in a pneumococcal population. Here, we show that this phasevarion generates multiple phenotypic differences relevant to pathobiology. Importantly, expression of conserved protein antigens varies with phasevarion switching. As capsule expression, a major pneumococcal virulence factor, is also controlled by the phasevarion, our work will inform the selection of the best candidates to include in a rationally designed, universal pneumococcal vaccine.
Collapse
|
50
|
Pereira JM, Xu S, Leong JM, Sousa S. The Yin and Yang of Pneumolysin During Pneumococcal Infection. Front Immunol 2022; 13:878244. [PMID: 35529870 PMCID: PMC9074694 DOI: 10.3389/fimmu.2022.878244] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/23/2022] [Indexed: 12/15/2022] Open
Abstract
Pneumolysin (PLY) is a pore-forming toxin produced by the human pathobiont Streptococcus pneumoniae, the major cause of pneumonia worldwide. PLY, a key pneumococcal virulence factor, can form transmembrane pores in host cells, disrupting plasma membrane integrity and deregulating cellular homeostasis. At lytic concentrations, PLY causes cell death. At sub-lytic concentrations, PLY triggers host cell survival pathways that cooperate to reseal the damaged plasma membrane and restore cell homeostasis. While PLY is generally considered a pivotal factor promoting S. pneumoniae colonization and survival, it is also a powerful trigger of the innate and adaptive host immune response against bacterial infection. The dichotomy of PLY as both a key bacterial virulence factor and a trigger for host immune modulation allows the toxin to display both "Yin" and "Yang" properties during infection, promoting disease by membrane perforation and activating inflammatory pathways, while also mitigating damage by triggering host cell repair and initiating anti-inflammatory responses. Due to its cytolytic activity and diverse immunomodulatory properties, PLY is integral to every stage of S. pneumoniae pathogenesis and may tip the balance towards either the pathogen or the host depending on the context of infection.
Collapse
Affiliation(s)
- Joana M. Pereira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Molecular and Cellular (MC) Biology PhD Program, ICBAS - Instituto de Ciência Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Shuying Xu
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, United States
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA, United States
| | - John M. Leong
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, United States
| | - Sandra Sousa
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| |
Collapse
|