1
|
Xu S, Tan S, Romanos P, Reedy JL, Zhang Y, Mansour MK, Vyas JM, Mecsas J, Mou H, Leong JM. Blocking HXA 3-mediated neutrophil elastase release during S. pneumoniae lung infection limits pulmonary epithelial barrier disruption and bacteremia. mBio 2024; 15:e0185624. [PMID: 39120139 PMCID: PMC11389395 DOI: 10.1128/mbio.01856-24] [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: 06/20/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Streptococcus pneumoniae (Sp), a leading cause of community-acquired pneumonia, can spread from the lung into the bloodstream to cause septicemia and meningitis, with a concomitant threefold increase in mortality. Limitations in vaccine efficacy and a rise in antimicrobial resistance have spurred searches for host-directed therapies that target pathogenic immune processes. Polymorphonuclear leukocytes (PMNs) are essential for infection control but can also promote tissue damage and pathogen spread. The major Sp virulence factor, pneumolysin, triggers acute inflammation by stimulating the 12-lipoxygenase (12-LOX) eicosanoid synthesis pathway in epithelial cells. This pathway is required for systemic spread in a mouse pneumonia model and produces a number of bioactive lipids, including hepoxilin A3 (HXA3), a hydroxy epoxide PMN chemoattractant that has been hypothesized to facilitate breach of mucosal barriers. To understand how 12-LOX-dependent inflammation promotes dissemination during Sp lung infection and dissemination, we utilized bronchial stem cell-derived air-liquid interface cultures that lack this enzyme to show that HXA3 methyl ester (HXA3-ME) is sufficient to promote basolateral-to-apical PMN transmigration, monolayer disruption, and concomitant Sp barrier breach. In contrast, PMN transmigration in response to the non-eicosanoid chemoattractant N-formyl-L-methionyl-L-leucyl-phenylalanine (fMLP) did not lead to epithelial disruption or bacterial translocation. Correspondingly, HXA3-ME but not fMLP increased the release of neutrophil elastase (NE) from Sp-infected PMNs. Pharmacologic blockade of NE secretion or activity diminished epithelial barrier disruption and bacteremia after pulmonary challenge of mice. Thus, HXA3 promotes barrier-disrupting PMN transmigration and NE release, pathological events that can be targeted to curtail systemic disease following pneumococcal pneumonia.IMPORTANCEStreptococcus pneumoniae (Sp), a leading cause of pneumonia, can spread from the lung into the bloodstream to cause systemic disease. Limitations in vaccine efficacy and a rise in antimicrobial resistance have spurred searches for host-directed therapies that limit pathologic host immune responses to Sp. Excessive polymorphonuclear leukocyte (PMN) infiltration into Sp-infected airways promotes systemic disease. Using stem cell-derived respiratory cultures that reflect bona fide lung epithelium, we identified eicosanoid hepoxilin A3 as a critical pulmonary PMN chemoattractant that is sufficient to drive PMN-mediated epithelial damage by inducing the release of neutrophil elastase. Inhibition of the release or activity of this protease in mice limited epithelial barrier disruption and bacterial dissemination, suggesting a new host-directed treatment for Sp lung infection.
Collapse
Affiliation(s)
- Shuying Xu
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, Massachusetts, USA
| | - Shumin Tan
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Patricia Romanos
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Program in Biotechnology, Francisco de Vitoria University, Madrid, Spain
| | - Jennifer L. Reedy
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Yihan Zhang
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michael K. Mansour
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jatin M. Vyas
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Joan Mecsas
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Hongmei Mou
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - John M. Leong
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Stuart B Levy Center for the Integrated Management of Antimicrobial Resistance, Tufts University, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Xu S, Tan S, Romanos P, Reedy JL, Zhang Y, Mansour MK, Vyas JM, Mecsas J, Mou H, Leong JM. Blocking HXA 3-mediated neutrophil elastase release during S. pneumoniae lung infection limits pulmonary epithelial barrier disruption and bacteremia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.25.600637. [PMID: 38979170 PMCID: PMC11230237 DOI: 10.1101/2024.06.25.600637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Streptococcus pneumoniae (Sp), a leading cause of community-acquired pneumonia, can spread from the lung into the bloodstream to cause septicemia and meningitis, with a concomitant three-fold increase in mortality. Limitations in vaccine efficacy and a rise in antimicrobial resistance have spurred searches for host-directed therapies that target pathogenic immune processes. Polymorphonuclear leukocytes (PMNs) are essential for infection control but can also promote tissue damage and pathogen spread. The major Sp virulence factor, pneumolysin (PLY), triggers acute inflammation by stimulating the 12-lipoxygenase (12-LOX) eicosanoid synthesis pathway in epithelial cells. This pathway is required for systemic spread in a mouse pneumonia model and produces a number of bioactive lipids, including hepoxilin A3 (HXA3), a hydroxy epoxide PMN chemoattractant that has been hypothesized to facilitate breach of mucosal barriers. To understand how 12-LOX-dependent inflammation promotes dissemination during Sp lung infection and dissemination, we utilized bronchial stem cell-derived air-liquid interface (ALI) cultures that lack this enzyme to show that HXA3 methyl ester (HXA3-ME) is sufficient to promote basolateral-to-apical PMN transmigration, monolayer disruption, and concomitant Sp barrier breach. In contrast, PMN transmigration in response to the non-eicosanoid chemoattractant fMLP did not lead to epithelial disruption or bacterial translocation. Correspondingly, HXA3-ME but not fMLP increased release of neutrophil elastase (NE) from Sp-infected PMNs. Pharmacologic blockade of NE secretion or activity diminished epithelial barrier disruption and bacteremia after pulmonary challenge of mice. Thus, HXA3 promotes barrier disrupting PMN transmigration and NE release, pathological events that can be targeted to curtail systemic disease following pneumococcal pneumonia.
Collapse
Affiliation(s)
- Shuying Xu
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA
| | - Shumin Tan
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA
| | - Patricia Romanos
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA
- Francisco de Vitoria University, Madrid, Spain
| | - Jennifer L. Reedy
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA
| | - Yihan Zhang
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA
| | - Michael K. Mansour
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA
| | - Jatin M. Vyas
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA
| | - Joan Mecsas
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA
| | - Hongmei Mou
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA
| | - John M. Leong
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA
- Stuart B Levy Center for the Integrated Management of Antimicrobial Resistance, Tufts University, Boston, MA
| |
Collapse
|
3
|
Sepahvand S, Madani M, Sepahvand H, Davarpanah MA. Comparative assessment of the mouse immune responses to colistin-resistant and colistin-sensitive isolates of Acinetobacter baumannii. Microb Pathog 2022; 173:105834. [DOI: 10.1016/j.micpath.2022.105834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/07/2022]
|
4
|
Immunogenicity and protective efficacy of a prototype pneumococcal bioconjugate vaccine. Vaccine 2022; 40:6107-6113. [PMID: 36115800 PMCID: PMC10388713 DOI: 10.1016/j.vaccine.2022.09.018] [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: 03/30/2022] [Revised: 08/01/2022] [Accepted: 09/05/2022] [Indexed: 11/22/2022]
Abstract
Capsular polysaccharides (CPSs), with which most pathogenic bacterial surfaces are decorated, have been used as the main components of glycoconjugate vaccines against bacterial diseases in clinical practice worldwide. Pneumococcal conjugate vaccines (PCVs) are administered globally to prevent invasive pneumococcal disease (IPD). While PCVs have played important roles in controlling IPD in all age groups, their empirical, and labor-intensive chemical conjugation yield poorly characterized, heterogeneous, and variably immunogenic vaccines, with poor immune responses in high-risk populations such as the elderly and patients with weak immune systems. We previously developed a method that bypasses the dependency of chemical conjugation and instead exploits prokaryotic glycosylation systems to produce pneumococcal conjugate vaccines. The bioconjugation platform relies on a conjugating enzyme to transfer a bacterial polysaccharide to an engineered carrier protein all within the lab safe bacterium E. coli. In these studies, we demonstrate that a serotype 8 pneumococcal bioconjugate vaccine is highly immunogenic and elicits functionally protective anti-serotype 8 antibody responses. Specifically, using multiple models we show that mice immunized with multiple doses of a serotype 8 bioconjugate vaccine elicit antibody responses that mediate opsonophagocytic killing, protect mice from systemic infection, and decrease the ability of serotype 8 pneumococci to colonize the nasopharynx and disseminate. Collectively, these studies demonstrate the utility of bioconjugation to produce efficacious pneumococcal conjugate vaccines.
Collapse
|
5
|
Extracellular Vesicles from Different Pneumococcal Serotypes Are Internalized by Macrophages and Induce Host Immune Responses. Pathogens 2021; 10:pathogens10121530. [PMID: 34959485 PMCID: PMC8708143 DOI: 10.3390/pathogens10121530] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/11/2022] Open
Abstract
Bacterial extracellular vesicles are membranous ultrastructures released from the cell surface. They play important roles in the interaction between the host and the bacteria. In this work, we show how extracellular vesicles produced by four different serotypes of the important human pathogen, Streptococcus pneumoniae, are internalized by murine J774A.1 macrophages via fusion with the membrane of the host cells. We also evaluated the capacity of pneumococcal extracellular vesicles to elicit an immune response by macrophages. Macrophages treated with the vesicles underwent a serotype-dependent transient loss of viability, which was further reverted. The vesicles induced the production of proinflammatory cytokines, which was higher for serotype 1 and serotype 8-derived vesicles. These results demonstrate the biological activity of extracellular vesicles of clinically important pneumococcal serotypes.
Collapse
|
6
|
Neutrophil-suppressive activity over T-cell proliferation and fungal clearance in a murine model of Fonsecaea pedrosoi infection. Sci Rep 2021; 11:20220. [PMID: 34642440 PMCID: PMC8511260 DOI: 10.1038/s41598-021-99847-z] [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: 05/17/2021] [Accepted: 09/15/2021] [Indexed: 11/28/2022] Open
Abstract
Neutrophils are essential to control several fungal infections. These cells are commonly known for their pro-inflammatory activities. However, some studies have demonstrated the anti-inflammatory properties of neutrophils during certain infectious diseases, culminating in the inhibition of T cell proliferation. Chromoblastomycosis (CBM) is a deep and progressive mycosis that affects thousands of people worldwide. Although neutrophil infiltrates are observed in the lesion histopathology, the fungus can overtake the immune system response and destroy the host-infected tissue. The present study demonstrated that neutropenic animals had an increase in the IL-6 production in the spleen and liver, followed by a lower fungal burden in these organs up to 14 days of infection. Neutropenic animals also showed a lower F. pedrosoi-specific antibody production 14-days post infection and higher T-cell proliferation in the in vitro experiments after stimulation with F. pedrosoi-purified proteins. Taken together, our results suggest that the presence of regulatory neutrophils in the mouse model of F. pedrosoi infection could act favoring the spread of the fungus and the chronicity of the infection. These findings shed light on the CBM treatment, which might target neutrophil polarization as a new therapy approach to treat CBM lesions.
Collapse
|
7
|
Anthony D, Papanicolaou A, Wang H, Seow HJ, To EE, Yatmaz S, Anderson GP, Wijburg O, Selemidis S, Vlahos R, Bozinovski S. Excessive Reactive Oxygen Species Inhibit IL-17A + γδ T Cells and Innate Cellular Responses to Bacterial Lung Infection. Antioxid Redox Signal 2020; 32:943-956. [PMID: 31190552 DOI: 10.1089/ars.2018.7716] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Aims: Excessive reactive oxygen species (ROS) are detrimental to immune cellular functions that control pathogenic microbes; however, the mechanisms are poorly understood. Our aim was to determine the immunological consequences of increased ROS levels during acute bacterial infection. Results: We used a model of Streptococcus pneumoniae (Spn) lung infection and superoxide dismutase 3-deficient (SOD3-/-) mice, as SOD3 is a major antioxidant enzyme that catalyses the dismutation of superoxide radicals. First, we observed that in vitro, macrophages from SOD3-/- mice generated excessive phagosomal ROS during acute bacterial infection. In vivo, there was a significant reduction in infiltrating neutrophils in the bronchoalveolar lavage fluid and reduced peribronchial and alveoli inflammation in SOD3-/- mice 2 days after Spn infection. Annexin V/propidium iodide staining revealed enhanced apoptosis in neutrophils from Spn-infected SOD3-/- mice. In addition, SOD3-/- mice showed an altered macrophage phenotypic profile, with markedly diminished recruitment of monocytes (CD11clo, CD11bhi) in the airways. Further investigation revealed significantly lower levels of the monocyte chemokine CCL-2, and cytokines IL-23, IL-1β, and IL-17A in Spn-infected SOD3-/- mice. There were also significantly fewer IL-17A-expressing gamma-delta T cells (γδ T cells) in the lungs of Spn-infected SOD3-/- mice. Innovation: Our data demonstrate that SOD3 deficiency leads to an accumulation of phagosomal ROS levels that initiate early neutrophil apoptosis during pneumococcal infection. Consequent to these events, there was a failure to initiate innate γδ T cell responses. Conclusion: These studies offer new cellular and mechanistic insights into how excessive ROS can regulate innate immune responses to bacterial infection.
Collapse
Affiliation(s)
- Desiree Anthony
- Program in Chronic Infectious and Inflammatory Diseases, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, Australia.,Department of Pharmacology & Therapeutics, Lung Health Research Centre, The University of Melbourne, Melbourne, Australia
| | - Angelica Papanicolaou
- Program in Chronic Infectious and Inflammatory Diseases, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, Australia
| | - Hao Wang
- Program in Chronic Infectious and Inflammatory Diseases, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, Australia
| | - Huei Jiunn Seow
- Program in Chronic Infectious and Inflammatory Diseases, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, Australia.,Department of Pharmacology & Therapeutics, Lung Health Research Centre, The University of Melbourne, Melbourne, Australia
| | - Eunice E To
- Program in Chronic Infectious and Inflammatory Diseases, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, Australia
| | - Selcuk Yatmaz
- Program in Chronic Infectious and Inflammatory Diseases, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, Australia
| | - Gary P Anderson
- Department of Pharmacology & Therapeutics, Lung Health Research Centre, The University of Melbourne, Melbourne, Australia
| | - Odilia Wijburg
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Australia
| | - Stavros Selemidis
- Program in Chronic Infectious and Inflammatory Diseases, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, Australia
| | - Ross Vlahos
- Program in Chronic Infectious and Inflammatory Diseases, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, Australia.,Department of Pharmacology & Therapeutics, Lung Health Research Centre, The University of Melbourne, Melbourne, Australia
| | - Steven Bozinovski
- Program in Chronic Infectious and Inflammatory Diseases, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, Australia.,Department of Pharmacology & Therapeutics, Lung Health Research Centre, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
8
|
Stackowicz J, Jönsson F, Reber LL. Mouse Models and Tools for the in vivo Study of Neutrophils. Front Immunol 2020; 10:3130. [PMID: 32038641 PMCID: PMC6985372 DOI: 10.3389/fimmu.2019.03130] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/23/2019] [Indexed: 12/21/2022] Open
Abstract
Neutrophils are the most abundant leukocytes in human blood and critical actors of the immune system. Many neutrophil functions and facets of their activity in vivo were revealed by studying genetically modified mice or by tracking fluorescent neutrophils in animals using imaging approaches. Assessing the roles of neutrophils can be challenging, especially when exact molecular pathways are questioned or disease states are interrogated that alter normal neutrophil homeostasis. This review discusses the main in vivo models for the study of neutrophils, their advantages and limitations. The side-by-side comparison underlines the necessity to carefully choose the right model(s) to answer a given scientific question, and exhibit caveats that need to be taken into account when designing experimental procedures. Collectively, this review suggests that at least two models should be employed to legitimately conclude on neutrophil functions.
Collapse
Affiliation(s)
- Julien Stackowicz
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France.,Sorbonne Université, Collège Doctoral, Paris, France
| | - Friederike Jönsson
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France
| | - Laurent L Reber
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France.,Center for Pathophysiology Toulouse-Purpan (CPTP), UMR 1043, University of Toulouse, INSERM, CNRS, Toulouse, France
| |
Collapse
|
9
|
G-CSFR antagonism reduces neutrophilic inflammation during pneumococcal and influenza respiratory infections without compromising clearance. Sci Rep 2019; 9:17732. [PMID: 31776393 PMCID: PMC6881371 DOI: 10.1038/s41598-019-54053-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 11/06/2019] [Indexed: 12/12/2022] Open
Abstract
Excessive neutrophilic inflammation can contribute to the pathogenesis of pneumonia. Whilst anti-inflammatory therapies such as corticosteroids are used to treat excessive inflammation, they do not selectively target neutrophils and may compromise antimicrobial or antiviral defences. In this study, neutrophil trafficking was targeted with a granulocyte-colony stimulating factor receptor monoclonal antibody (G-CSFR mAb) during Streptococcus pneumoniae (serotype 19F) or influenza A virus (IAV, strain HKx31) lung infection in mice. Firstly, we demonstrated that neutrophils are indispensable for the clearance of S. pneumoniae from the airways using an anti-Ly6G monoclonal antibody (1A8 mAb), as the complete inhibition of neutrophil recruitment markedly compromised bacterial clearance. Secondly, we demonstrated that G-CSF transcript lung levels were significantly increased during pneumococcal infection. Prophylactic or therapeutic administration of G-CSFR mAb significantly reduced blood and airway neutrophil numbers by 30–60% without affecting bacterial clearance. Total protein levels in the bronchoalveolar lavage (BAL) fluid (marker for oedema) was also significantly reduced. G-CSF transcript levels were also increased during IAV lung infection. G-CSFR mAb treatment significantly reduced neutrophil trafficking into BAL compartment by 60% and reduced blood neutrophil numbers to control levels in IAV-infected mice. Peak lung viral levels at day 3 were not altered by G-CSFR therapy, however there was a significant reduction in the detection of IAV in the lungs at the day 7 post-infection phase. In summary, G-CSFR signalling contributes to neutrophil trafficking in response to two common respiratory pathogens. Blocking G-CSFR reduced neutrophil trafficking and oedema without compromising clearance of two pathogens that can cause pneumonia.
Collapse
|
10
|
Age-related changes in the levels and kinetics of pulmonary cytokine and chemokine responses to Streptococcuspneumoniae in mouse pneumonia models. Cytokine 2018; 111:389-397. [PMID: 30463053 DOI: 10.1016/j.cyto.2018.09.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 11/20/2022]
Abstract
Streptococcuspneumoniae is a major human pathogen at the extremes of age. The elderly are particularly vulnerable to S.pneumoniae, the most common causative agent of bacterial pneumonia in this population. Despite the availability of vaccines and antibiotics, mortality rates associated with pneumococcal pneumonia in this age group remain high. In light of globally increasing life-expectancy, a better understanding of the patho-mechanisms of elderly pneumococcal pneumonia, including alterations in innate immune responses, is needed to develop improved therapies. In this study we aimed at investigating how increased susceptibility to pneumococcal infection relates to inflammation kinetics in the aged mouse pneumonia model by determining pulmonary cytokine and chemokine levels and comparing these parameters to those measured in young adult mice. Firstly, we detected overall higher pulmonary cytokine and chemokine levels in aged mice. However, upon induction of pneumococcal pneumonia in aged mice, delayed production of certain analytes, such as IFN-γ, MIG (CXCL9), IP-10 (CXCL10), MCP-1 (CCL2), TARC (CCL17) and MDC (CCL22) became apparent. In addition, aged mice were unable to control excess inflammatory responses: while young mice showed peak inflammatory responses at 20 h and subsequent resolution by 48 h post intranasal challenge, in aged mice increasing cytokine and chemokine levels were measured. These findings highlight the importance of considering multiple time points when delineating inflammatory responses to S.pneumoniae in an age-related context. Finally, correlation between pulmonary bacterial burden and cytokine or chemokine levels in young mice suggested that appropriately controlled inflammatory responses support the host to fight pneumococcal infection.
Collapse
|
11
|
Transporters MRP1 and MRP2 Regulate Opposing Inflammatory Signals To Control Transepithelial Neutrophil Migration during Streptococcus pneumoniae Lung Infection. mSphere 2018; 3:3/4/e00303-18. [PMID: 29976647 PMCID: PMC6034076 DOI: 10.1128/msphere.00303-18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Streptococcus pneumoniae is a Gram-positive bacterium that normally inhabits the human nasopharynx asymptomatically. However, it is also a major cause of pneumonia, bacteremia, and meningitis. The transition from pneumonia to bacteremia is critical, as patients that develop septicemia have ~20% mortality rates. Previous studies have shown that while neutrophils, a major bacterium-induced leukocyte, aid in S. pneumoniae elimination, they also contribute to pathology and may mediate the lung-to-blood passage of the bacteria. Herein, we show that epithelium-derived MRP1 and MRP2 efflux immunomodulatory agents that assist in controlling passage of neutrophils during infection and that limiting neutrophil infiltration produced less bacteremia and better survival during murine infection. The importance of our work is twofold: ours is the first to identify an MRP1/MRP2 axis of neutrophil control in the lung. The second is to provide possible therapeutic targets to reduce excess inflammation, thus reducing the chances of developing bacteremia during pneumococcal pneumonia. Streptococcus pneumoniae remains a source of morbidity and mortality in both developed and underdeveloped nations of the world. Disease can manifest as pneumonia, bacteremia, and meningitis, depending on the localization of infection. Interestingly, there is a correlation in experimental murine infections between the development of bacteremia and influx of neutrophils into the pulmonary lumen. Reduction of this neutrophil influx has been shown to improve survivability during infection. In this study, we use in vitro biotinylation and neutrophil transmigration and in vivo murine infection to identify a system in which two epithelium-localized ATP-binding cassette transporters, MRP1 and MRP2, have inverse activities dictating neutrophil transmigration into the lumen of infected mouse lungs. MRP1 effluxes an anti-inflammatory molecule that maintains homeostasis in uninfected contexts, thus reducing neutrophil infiltration. During inflammatory events, however, MRP1 decreases and MRP2 both increases and effluxes the proinflammatory eicosanoid hepoxilin A3. If we then decrease MRP2 activity during experimental murine infection with S. pneumoniae, we reduce both neutrophil infiltration and bacteremia, showing that MRP2 coordinates this activity in the lung. We conclude that MRP1 assists in depression of polymorphonuclear cell (PMN) migration by effluxing a molecule that inhibits the proinflammatory effects of MRP2 activity. IMPORTANCEStreptococcus pneumoniae is a Gram-positive bacterium that normally inhabits the human nasopharynx asymptomatically. However, it is also a major cause of pneumonia, bacteremia, and meningitis. The transition from pneumonia to bacteremia is critical, as patients that develop septicemia have ~20% mortality rates. Previous studies have shown that while neutrophils, a major bacterium-induced leukocyte, aid in S. pneumoniae elimination, they also contribute to pathology and may mediate the lung-to-blood passage of the bacteria. Herein, we show that epithelium-derived MRP1 and MRP2 efflux immunomodulatory agents that assist in controlling passage of neutrophils during infection and that limiting neutrophil infiltration produced less bacteremia and better survival during murine infection. The importance of our work is twofold: ours is the first to identify an MRP1/MRP2 axis of neutrophil control in the lung. The second is to provide possible therapeutic targets to reduce excess inflammation, thus reducing the chances of developing bacteremia during pneumococcal pneumonia.
Collapse
|
12
|
Ritchie ND, Ritchie R, Bayes HK, Mitchell TJ, Evans TJ. IL-17 can be protective or deleterious in murine pneumococcal pneumonia. PLoS Pathog 2018; 14:e1007099. [PMID: 29813133 PMCID: PMC5993294 DOI: 10.1371/journal.ppat.1007099] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/08/2018] [Accepted: 05/14/2018] [Indexed: 12/21/2022] Open
Abstract
Streptococcus pneumoniae is the major bacterial cause of community-acquired pneumonia, and the leading agent of childhood pneumonia deaths worldwide. Nasal colonization is an essential step prior to infection. The cytokine IL-17 protects against such colonization and vaccines that enhance IL-17 responses to pneumococcal colonization are being developed. The role of IL-17 in host defence against pneumonia is not known. To address this issue, we have utilized a murine model of pneumococcal pneumonia in which the gene for the IL-17 cytokine family receptor, Il17ra, has been inactivated. Using this model, we show that IL-17 produced predominantly from γδ T cells protects mice against death from the invasive TIGR4 strain (serotype 4) which expresses a relatively thin capsule. However, in pneumonia produced by two heavily encapsulated strains with low invasive potential (serotypes 3 and 6B), IL-17 significantly enhanced mortality. Neutrophil uptake and killing of the serotype 3 strain was significantly impaired compared to the serotype 4 strain and depletion of neutrophils with antibody enhanced survival of mice infected with the highly encapsulated SRL1 strain. These data strongly suggest that IL-17 mediated neutrophil recruitment to the lungs clears infection from the invasive TIGR4 strain but that lung neutrophils exacerbate disease caused by the highly encapsulated pneumococcal strains. Thus, whilst augmenting IL-17 immune responses against pneumococci may decrease nasal colonization, this may worsen outcome during pneumonia caused by some strains.
Collapse
MESH Headings
- Animals
- Bacteremia/immunology
- Bacteremia/microbiology
- Bacterial Capsules/immunology
- Bacterial Capsules/ultrastructure
- Bronchoalveolar Lavage Fluid/cytology
- Bronchoalveolar Lavage Fluid/microbiology
- Disease Models, Animal
- Interleukin-17/immunology
- Lung/cytology
- Lung/enzymology
- Lung/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Electron, Transmission
- Microscopy, Fluorescence
- Nasopharynx/microbiology
- Neutrophils/cytology
- Neutrophils/immunology
- Peroxidase/metabolism
- Phagocytosis
- Pneumonia, Pneumococcal/immunology
- Pneumonia, Pneumococcal/mortality
- Pneumonia, Pneumococcal/prevention & control
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Interleukin-17/genetics
- Specific Pathogen-Free Organisms
- Streptococcus pneumoniae/immunology
- Streptococcus pneumoniae/ultrastructure
Collapse
Affiliation(s)
- Neil D. Ritchie
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Ryan Ritchie
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Hannah K. Bayes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Tim J. Mitchell
- Institute of Microbiology and Infection, College of Medical and Dental Sciences University of Birmingham, Birmingham, United Kingdom
| | - Tom J. Evans
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
13
|
Cytosolic Phospholipase A 2α Promotes Pulmonary Inflammation and Systemic Disease during Streptococcus pneumoniae Infection. Infect Immun 2017; 85:IAI.00280-17. [PMID: 28808157 DOI: 10.1128/iai.00280-17] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/02/2017] [Indexed: 02/07/2023] Open
Abstract
Pulmonary infection by Streptococcus pneumoniae is characterized by a robust alveolar infiltration of neutrophils (polymorphonuclear cells [PMNs]) that can promote systemic spread of the infection if not resolved. We previously showed that 12-lipoxygenase (12-LOX), which is required to generate the PMN chemoattractant hepoxilin A3 (HXA3) from arachidonic acid (AA), promotes acute pulmonary inflammation and systemic infection after lung challenge with S. pneumoniae As phospholipase A2 (PLA2) promotes the release of AA, we investigated the role of PLA2 in local and systemic disease during S. pneumoniae infection. The group IVA cytosolic isoform of PLA2 (cPLA2α) was activated upon S. pneumoniae infection of cultured lung epithelial cells and was critical for AA release from membrane phospholipids. Pharmacological inhibition of this enzyme blocked S. pneumoniae-induced PMN transepithelial migration in vitro Genetic ablation of the cPLA2 isoform cPLA2α dramatically reduced lung inflammation in mice upon high-dose pulmonary challenge with S. pneumoniae The cPLA2α-deficient mice also suffered no bacteremia and survived a pulmonary challenge that was lethal to wild-type mice. Our data suggest that cPLA2α plays a crucial role in eliciting pulmonary inflammation during pneumococcal infection and is required for lethal systemic infection following S. pneumoniae lung challenge.
Collapse
|
14
|
THP-1-derived macrophages render lung epithelial cells hypo-responsive to Legionella pneumophila - a systems biology study. Sci Rep 2017; 7:11988. [PMID: 28931863 PMCID: PMC5607273 DOI: 10.1038/s41598-017-12154-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 09/04/2017] [Indexed: 11/25/2022] Open
Abstract
Immune response in the lung has to protect the huge alveolar surface against pathogens while securing the delicate lung structure. Macrophages and alveolar epithelial cells constitute the first line of defense and together orchestrate the initial steps of host defense. In this study, we analysed the influence of macrophages on type II alveolar epithelial cells during Legionella pneumophila-infection by a systems biology approach combining experimental work and mathematical modelling. We found that L. pneumophila-infected THP-1-derived macrophages provoke a pro-inflammatory activation of neighboring lung epithelial cells, but in addition render them hypo-responsive to direct infection with the same pathogen. We generated a kinetic mathematical model of macrophage activation and identified a paracrine mechanism of macrophage-secreted IL-1β inducing a prolonged IRAK-1 degradation in lung epithelial cells. This intercellular crosstalk may help to avoid an overwhelming inflammatory response by preventing excessive local secretion of pro-inflammatory cytokines and thereby negatively regulating the recruitment of immune cells to the site of infection. This suggests an important but ambivalent immunomodulatory role of macrophages in lung infection.
Collapse
|
15
|
Zou J, Zhou L, Hu C, Jing P, Guo X, Liu S, Lei Y, Yang S, Deng J, Zhang H. IL-8 and IP-10 expression from human bronchial epithelial cells BEAS-2B are promoted by Streptococcus pneumoniae endopeptidase O (PepO). BMC Microbiol 2017; 17:187. [PMID: 28836948 PMCID: PMC5571634 DOI: 10.1186/s12866-017-1081-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/31/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The bronchial epithelium serves as the first defendant line of host against respiratory inhaled pathogens, mainly through releasing chemokines (e.g. interleukin-8 (IL-8), interferon-induced protein 10 (IP-10) etc.) responsible for neutrophil or lymphocyte recruitment to promote the clearance of inhaled pathogens including Streptococcus pneumoniae (S. pneumoniae). Previous studies have shown that IL-8 expression is induced by pneumococcal virulence factors (e.g. pneumolysin, peptidoglycan-polysaccharides, pneumococcal surface protein A (PspA) etc.), which contributes to the pathogenesis of pneumonia. Whether other pneumococcal virulence factors are involved in inducing chemokines expression in epithelium is still unknown. RESULTS We studied the effect of PepO, a widely expressed and newly discovered pneumococcal virulence protein, on the release of proinflammatory cytokines, IL-8 and IP-10, from human bronchial epithelial cell line BEAS-2B and identified the relevant signaling pathways. Incubation of BEAS-2B with PepO resulted in increased synthesis and release of IL-8 and IP-10 in a dose and time independent manner. We also detected the increased and sustained expression of TLR2 and TLR4 transcripts in BEAS-2B stimulated by PepO. PepO activation leaded to the phosphorylation of MAPKs, Akt and p65. Pharmacologic inhibitors of MAPKs, PI3K and IκB-α phosphorylation attenuated IL-8 release, while IP-10 production was just suppressed by inhibitors of IκB-α phosphorylation, PI3K and P38 MAPK. CONCLUSION These results suggest that PepO enhances IL-8 and IP-10 production in BEAS-2B in a MAPKs-PI3K/Akt-p65 dependent manner, which may play critical roles in the pathogenesis of pneumonia.
Collapse
Affiliation(s)
- Jiaqiong Zou
- Department of Laboratory Medicine, The Affiliated Hospital of North Sichuan Medical College, 63 Wenhua Road, Shunqing District, Nanchong, Sichuan, 637000, China.,Department of Laboratory Medicine, North Sichuan Medical College; Translational Medicine Research Center, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Long Zhou
- Department of Laboratory Medicine, Chongqing Three Gorges Central Hospital, Wanzhou, Chongqing, 404100, China
| | - Chunlan Hu
- Department of General Medicine, Chongqing Three Gorges Central Hospital, Wanzhou, Chongqing, 404100, China
| | - Peng Jing
- Department of Laboratory Medicine, The Affiliated Hospital of North Sichuan Medical College, 63 Wenhua Road, Shunqing District, Nanchong, Sichuan, 637000, China.,Department of Laboratory Medicine, North Sichuan Medical College; Translational Medicine Research Center, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xiaolan Guo
- Department of Pediatric Surgery, The Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Sulan Liu
- Department of Laboratory Medicine, The Affiliated Hospital of North Sichuan Medical College, 63 Wenhua Road, Shunqing District, Nanchong, Sichuan, 637000, China.,Department of Laboratory Medicine, North Sichuan Medical College; Translational Medicine Research Center, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yan Lei
- Department of Laboratory Medicine, The Affiliated Hospital of North Sichuan Medical College, 63 Wenhua Road, Shunqing District, Nanchong, Sichuan, 637000, China.,Department of Laboratory Medicine, North Sichuan Medical College; Translational Medicine Research Center, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Shangyu Yang
- Department of Laboratory Medicine, The Affiliated Hospital of North Sichuan Medical College, 63 Wenhua Road, Shunqing District, Nanchong, Sichuan, 637000, China.,Department of Laboratory Medicine, North Sichuan Medical College; Translational Medicine Research Center, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jiankang Deng
- Department of Laboratory Medicine, The Affiliated Hospital of North Sichuan Medical College, 63 Wenhua Road, Shunqing District, Nanchong, Sichuan, 637000, China. .,Department of Laboratory Medicine, North Sichuan Medical College; Translational Medicine Research Center, North Sichuan Medical College, Nanchong, Sichuan, China.
| | - Hong Zhang
- Department of Laboratory Medicine, The Affiliated Hospital of North Sichuan Medical College, 63 Wenhua Road, Shunqing District, Nanchong, Sichuan, 637000, China. .,Department of Laboratory Medicine, North Sichuan Medical College; Translational Medicine Research Center, North Sichuan Medical College, Nanchong, Sichuan, China.
| |
Collapse
|
16
|
Contribution of the Ade Resistance-Nodulation-Cell Division-Type Efflux Pumps to Fitness and Pathogenesis of Acinetobacter baumannii. mBio 2016; 7:mBio.00697-16. [PMID: 27247231 PMCID: PMC4895114 DOI: 10.1128/mbio.00697-16] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Overexpression of chromosomal resistance-nodulation-cell division (RND)-type efflux systems with broad substrate specificity contributes to multidrug resistance (MDR) in Acinetobacter baumannii. We have shown that modulation of expression of the structural genes for the efflux systems AdeABC and AdeIJK confers MDR and results in numerous alterations of membrane-associated cellular functions, in particular biofilm formation. However, the contribution of these RND pumps to cell fitness and virulence has not yet been studied. The biological cost of an antibiotic resistance mechanism is a key parameter in determining its stability and dissemination. From an entirely sequenced susceptible clinical isolate, we have generated a set of isogenic derivatives having single point mutations resulting in overexpression of each efflux system or with every pump deleted by allelic replacement. We found that overproduction of the pumps results in a significant decrease in fitness of the bacterial host when measured by competition experiments in vitro. Fitness and virulence were also evaluated in vivo both in systemic and pulmonary infection models in immunocompetent mice. A diminished competitiveness of the AdeABC-overexpressing mutant was observed only after intraperitoneal inoculation, but not after intranasal inoculation, the latter mimicking the most frequent type of human A. baumannii infection. However, in mice infected intranasally, this mutant was more virulent and stimulated an enhanced neutrophil activation in the lungs. Altogether, these data account for the observation that adeABC overexpression is common in MDR A. baumannii frequently found in ventilator-associated pneumonia. Overproduction of the RND AdeABC efflux system is observed with a high incidence in multidrug-resistant Acinetobacter baumannii and results in increased resistance to several antibiotics of choice for the treatment of infections caused by this nosocomial pathogen. It was therefore important to study the biological cost of the overexpression of the adeABC structural operon which is normally tightly regulated. Fitness diminution of an overexpressing mutant detected in vitro and in vivo in a model that mimics sepsis was not observed in a pulmonary infection model in which the mutant was more virulent. This points out that increased virulence can occur independently from prolonged persistence in the infected organ and can account for the elevated incidence of this resistance mechanism in clinical isolates. The study also indicates that transposon libraries will identify only virulence genes that are expressed under physiological conditions but not those that are tightly regulated.
Collapse
|
17
|
Immune ageing and susceptibility to Streptococcus pneumoniae. Biogerontology 2015; 17:449-65. [DOI: 10.1007/s10522-015-9614-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 10/05/2015] [Indexed: 12/16/2022]
|
18
|
Bou Ghanem EN, Clark S, Roggensack SE, McIver SR, Alcaide P, Haydon PG, Leong JM. Extracellular Adenosine Protects against Streptococcus pneumoniae Lung Infection by Regulating Pulmonary Neutrophil Recruitment. PLoS Pathog 2015; 11:e1005126. [PMID: 26313746 PMCID: PMC4552087 DOI: 10.1371/journal.ppat.1005126] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 08/04/2015] [Indexed: 12/15/2022] Open
Abstract
An important determinant of disease following Streptococcus pneumoniae (pneumococcus) lung infection is pulmonary inflammation mediated by polymorphonuclear leukocytes (PMNs). We found that upon intratracheal challenge of mice, recruitment of PMNs into the lungs within the first 3 hours coincided with decreased pulmonary pneumococci, whereas large numbers of pulmonary PMNs beyond 12 hours correlated with a greater bacterial burden. Indeed, mice that survived infection largely resolved inflammation by 72 hours, and PMN depletion at peak infiltration, i.e. 18 hours post-infection, lowered bacterial numbers and enhanced survival. We investigated host signaling pathways that influence both pneumococcus clearance and pulmonary inflammation. Pharmacologic inhibition and/or genetic ablation of enzymes that generate extracellular adenosine (EAD) (e.g. the ectoenzyme CD73) or degrade EAD (e.g. adenosine deaminase) revealed that EAD dramatically increases murine resistance to S. pneumoniae lung infection. Moreover, adenosine diminished PMN movement across endothelial monolayers in vitro, and although inhibition or deficiency of CD73 had no discernible impact on PMN recruitment within the first 6 hours after intratracheal inoculation of mice, these measures enhanced PMN numbers in the pulmonary interstitium after 18 hours of infection, culminating in dramatically elevated numbers of pulmonary PMNs at three days post-infection. When assessed at this time point, CD73-/- mice displayed increased levels of cellular factors that promote leukocyte migration, such as CXCL2 chemokine in the murine lung, as well as CXCR2 and β-2 integrin on the surface of pulmonary PMNs. The enhanced pneumococcal susceptibility of CD73-/- mice was significantly reversed by PMN depletion following infection, suggesting that EAD-mediated resistance is largely mediated by its effects on PMNs. Finally, CD73-inhibition diminished the ability of PMNs to kill pneumococci in vitro, suggesting that EAD alters both the recruitment and bacteriocidal function of PMNs. The EAD-pathway may provide a therapeutic target for regulating potentially harmful inflammatory host responses during Gram-positive bacterial pneumonia. Despite the presence of vaccines and antibiotic therapies, invasive Streptococcus pneumoniae (pneumococcus) infections, such as pneumonia, bacteremia and meningitis, remain a leading cause of mortality and morbidity worldwide. Understanding the host factors that influence the outcome of S. pneumoniae infection will allow us to design better therapies. Here, we elucidate the role of rapidly responding innate immune cells termed neutrophils, or PMNs (polymorphonuclear leukocytes), whose role in S. pneumoniae infection has long been controversial. We found that PMNs are initially required for controlling bacterial numbers, but their extended presence in the lungs leads to significant damage and poor control of infection. The signals that control the movement of PMNs into the infected lungs are not well understood. Here, we identified extracellular adenosine (EAD), a molecule produced by the host in response to cellular damage, as important in limiting PMN movement into the lungs upon pneumococcal challenge. Importantly, EAD-mediated control of PMNs was crucial for fighting lung infection by S. pneumoniae. This study may lead to the potential use of clinically available adenosine-based therapies to combat pneumococcal pneumonia in the future.
Collapse
Affiliation(s)
- Elsa N. Bou Ghanem
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Stacie Clark
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Sara E. Roggensack
- Program in Molecular Microbiology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts, United States of America
| | - Sally R. McIver
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Pilar Alcaide
- Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine and Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Philip G. Haydon
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - John M. Leong
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
19
|
Sanfilippo AM, Furuya Y, Roberts S, Salmon SL, Metzger DW. Allergic Lung Inflammation Reduces Tissue Invasion and Enhances Survival from Pulmonary Pneumococcal Infection in Mice, Which Correlates with Increased Expression of Transforming Growth Factor β1 and SiglecF(low) Alveolar Macrophages. Infect Immun 2015; 83:2976-83. [PMID: 25964474 PMCID: PMC4468552 DOI: 10.1128/iai.00142-15] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 05/04/2015] [Indexed: 11/20/2022] Open
Abstract
Asthma is generally thought to confer an increased risk for invasive pneumococcal disease (IPD) in humans. However, recent reports suggest that mortality rates from IPD are unaffected in patients with asthma and that chronic obstructive pulmonary disease (COPD), a condition similar to asthma, protects against the development of complicated pneumonia. To clarify the effects of asthma on the subsequent susceptibility to pneumococcal infection, ovalbumin (OVA)-induced allergic lung inflammation (ALI) was induced in mice followed by intranasal infection with A66.1 serotype 3 Streptococcus pneumoniae. Surprisingly, mice with ALI were significantly more resistant to lethal infection than non-ALI mice. The heightened resistance observed following ALI correlated with enhanced early clearance of pneumococci from the lung, decreased bacterial invasion from the airway into the lung tissue, a blunted inflammatory cytokine and neutrophil response to infection, and enhanced expression of transforming growth factor β1 (TGF-β1). Neutrophil depletion prior to infection had no effect on enhanced early bacterial clearance or resistance to IPD in mice with ALI. Although eosinophils recruited into the lung during ALI appeared to be capable of phagocytizing bacteria, neutralization of interleukin-5 (IL-5) to inhibit eosinophil recruitment likewise had no effect on early clearance or survival following infection. However, enhanced resistance was associated with an increase in levels of clodronate-sensitive, phagocytic SiglecF(low) alveolar macrophages within the airways following ALI. These findings suggest that, while the risk of developing IPD may actually be decreased in patients with acute asthma, additional clinical data are needed to better understand the risk of IPD in patients with different asthma phenotypes.
Collapse
Affiliation(s)
- Alan M Sanfilippo
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Yoichi Furuya
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Sean Roberts
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Sharon L Salmon
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Dennis W Metzger
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| |
Collapse
|
20
|
Weiser JN, Roche AM, Hergott CB, LaRose MI, Connolly T, Jorgensen WL, Leng L, Bucala R, Das R. Macrophage Migration Inhibitory Factor Is Detrimental in Pneumococcal Pneumonia and a Target for Therapeutic Immunomodulation. J Infect Dis 2015; 212:1677-82. [PMID: 25943202 DOI: 10.1093/infdis/jiv262] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 04/22/2015] [Indexed: 01/01/2023] Open
Abstract
Mortality from pneumococcal pneumonia remains high despite antibiotic therapy, highlighting the pathogenic potential for host inflammation. We demonstrate that macrophage migration inhibitory factor (MIF), an innate immune mediator, is detrimental for survival and associated with lung pathology, inflammatory cellular infiltration, and bacterial replication in a mouse model of pneumococcal pneumonia, despite being necessary for clearance from the nasopharynx. Treatment of animals with a small-molecule inhibitor of MIF improves survival by reducing inflammation and improving bacterial control. Our work demonstrates that MIF modulates beneficial versus detrimental inflammatory responses in the host-pneumococcal interaction and is a potential target for therapeutic modulation.
Collapse
Affiliation(s)
| | | | - Christopher B Hergott
- Department of Microbiology Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Meredith I LaRose
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | | | | | - Lin Leng
- Department of Medicine, Yale School of Medicine
| | | | - Rituparna Das
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| |
Collapse
|
21
|
Bou Ghanem EN, Clark S, Du X, Wu D, Camilli A, Leong JM, Meydani SN. The α-tocopherol form of vitamin E reverses age-associated susceptibility to streptococcus pneumoniae lung infection by modulating pulmonary neutrophil recruitment. THE JOURNAL OF IMMUNOLOGY 2014; 194:1090-9. [PMID: 25512603 PMCID: PMC4834212 DOI: 10.4049/jimmunol.1402401] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Streptococcus pneumoniae infections are an important cause of morbidity and mortality in older patients. Uncontrolled neutrophil-driven pulmonary inflammation exacerbates this disease. To test whether the α-tocopherol (α-Toc) form of vitamin E, a regulator of immunity, can modulate neutrophil responses as a preventive strategy to mitigate the age-associated decline in resistance to S. pneumoniae, young (4 mo) and old (22-24 mo) C57BL/6 mice were fed a diet containing 30-PPM (control) or 500-PPM (supplemented) α-Toc for 4 wk and intratracheally infected with S. pneumoniae. Aged mice fed a control diet were exquisitely more susceptible to S. pneumoniae than young mice. At 2 d postinfection, aged mice suffered 1000-fold higher pulmonary bacterial burden, 2.2-fold higher levels of neutrophil recruitment to the lung, and a 2.25-fold higher rate of lethal septicemia. Strikingly, α-Toc supplementation of aged mice resulted in a 1000-fold lower bacterial lung burden and full control of infection. This α-Toc-induced resistance to pneumococcal challenge was associated with a 2-fold fewer pulmonary neutrophils, a level comparable to S. pneumoniae-challenged, conventionally fed young mice. α-Toc directly inhibited neutrophil egress across epithelial cell monolayers in vitro in response to pneumococci or hepoxilin-A3, an eicosanoid required for pneumococcus-elicited neutrophil trans-epithelial migration. α-Toc altered expression of multiple epithelial and neutrophil adhesion molecules involved in migration, including CD55, CD47, CD18/CD11b, and ICAM-1. These findings suggest that α-Toc enhances resistance of aged mice to bacterial pneumonia by modulating the innate immune response, a finding that has potential clinical significance in combating infection in aged individuals through nutritional intervention.
Collapse
Affiliation(s)
- Elsa N Bou Ghanem
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111
| | - Stacie Clark
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111
| | - Xiaogang Du
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston MA 02114; and
| | - Dayong Wu
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston MA 02114; and
| | - Andrew Camilli
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111; Howard Hughes Medical Institute, Boston, MA 02111
| | - John M Leong
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111;
| | - Simin N Meydani
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston MA 02114; and
| |
Collapse
|
22
|
KuoLee R, Harris G, Yan H, Xu HH, Conlan WJ, Patel GB, Chen W. Intranasal immunization protects against Acinetobacter baumannii-associated pneumonia in mice. Vaccine 2014; 33:260-7. [PMID: 24699469 DOI: 10.1016/j.vaccine.2014.02.083] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Revised: 01/24/2014] [Accepted: 02/25/2014] [Indexed: 10/25/2022]
Abstract
Multidrug-resistant Acinetobacter baumannii has become an important causative agent of healthcare associated infections. Hospital- and community-acquired pneumonia is the most common clinical manifestation of A. baumannii infection worldwide and is often associated with high mortality. Most experimental vaccine studies to date have evaluated vaccines against systemic A. baumannii infections following systemic immunization. We recently demonstrated that a mouse model of respiratory A. baumannii infection using the strain LAC-4 results in disease progression that is similar to that observed in humans. Here we used this model in conjunction with an inactivated whole cell vaccine to evaluate the feasibility of developing protective mucosal vaccines against respiratory A. baumannii infection and to investigate the potential mechanism of protection of such vaccines. Our results showed that intranasal immunization with formalin-killed whole cells of the LAC-4 strain elicited mucosal and systemic antigen-specific immune responses, and protected mice against lethal intranasal or intraperitoneal challenges. Compared to naïve mice, immunized mice had significantly fewer bacteria in their lungs, and the pathogen was barely detectable in blood and spleens at 24h post challenge, indicating the ability of immunized mice to control extrapulmonary dissemination of the pathogen. Mechanistic studies using gene-deficient mice, neutropenic mice, or passive immunization showed that B cells and neutrophils, but not FcRγ, played crucial roles in the protection against respiratory A. baumannii challenge of intranasally immunized mice whereas passive transfer of hyperimmune sera only prolonged the survival time of challenged mice by 48 h. These results provide immunological insights for the rational design of novel mucosal vaccines to protect against respiratory A. baumannii infection and demonstrate the feasibility to develop such vaccines.
Collapse
Affiliation(s)
- Rhonda KuoLee
- Human Health Therapeutics, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada
| | - Greg Harris
- Human Health Therapeutics, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada
| | - Hongbin Yan
- Department of Chemistry, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - H Howard Xu
- Department of Biological Sciences, California State University, Los Angeles, Los Angeles, CA 90032, USA
| | - Wayne J Conlan
- Human Health Therapeutics, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada
| | - Girishchandra B Patel
- Human Health Therapeutics, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada
| | - Wangxue Chen
- Human Health Therapeutics, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada; Department of Biology, Brock University, St. Catharines, Ontario L2S 3A1, Canada.
| |
Collapse
|
23
|
Tailoring the Immune Response via Customization of Pathogen Gene Expression. J Pathog 2014; 2014:651568. [PMID: 24719769 PMCID: PMC3955589 DOI: 10.1155/2014/651568] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 12/23/2013] [Indexed: 12/27/2022] Open
Abstract
The majority of studies focused on the construction and reengineering of bacterial pathogens have mainly relied on the knocking out of virulence factors or deletion/mutation of amino acid residues to then observe the microbe's phenotype and the resulting effect on the host immune response. These knockout bacterial strains have also been proposed as vaccines to combat bacterial disease. Theoretically, knockout strains would be unable to cause disease since their virulence factors have been removed, yet they could induce a protective memory response. While knockout strains have been valuable tools to discern the role of virulence factors in host immunity and bacterial pathogenesis, they have been unable to yield clinically relevant vaccines. The advent of synthetic biology and enhanced user-directed gene customization has altered this binary process of knockout, followed by observation. Recent studies have shown that a researcher can now tailor and customize a given microbe's gene expression to produce a desired immune response. In this commentary, we highlight these studies as a new avenue for controlling the inflammatory response as well as vaccine development.
Collapse
|
24
|
Bhowmick R, Maung N, Hurley BP, Ghanem EB, Gronert K, McCormick BA, Leong JM. Systemic disease during Streptococcus pneumoniae acute lung infection requires 12-lipoxygenase-dependent inflammation. THE JOURNAL OF IMMUNOLOGY 2013; 191:5115-23. [PMID: 24089193 DOI: 10.4049/jimmunol.1300522] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Acute pulmonary infection by Streptococcus pneumoniae is characterized by high bacterial numbers in the lung, a robust alveolar influx of polymorphonuclear cells (PMNs), and a risk of systemic spread of the bacterium. We investigated host mediators of S. pneumoniae-induced PMN migration and the role of inflammation in septicemia following pneumococcal lung infection. Hepoxilin A3 (HXA3) is a PMN chemoattractant and a metabolite of the 12-lipoxygenase (12-LOX) pathway. We observed that S. pneumoniae infection induced the production of 12-LOX in cultured pulmonary epithelium and in the lungs of infected mice. Inhibition of the 12-LOX pathway prevented pathogen-induced PMN transepithelial migration in vitro and dramatically reduced lung inflammation upon high-dose pulmonary challenge with S. pneumoniae in vivo, thus implicating HXA3 in pneumococcus-induced pulmonary inflammation. PMN basolateral-to-apical transmigration in vitro significantly increased apical-to-basolateral transepithelial migration of bacteria. Mice suppressed in the expression of 12-LOX exhibited little or no bacteremia and survived an otherwise lethal pulmonary challenge. Our data suggest that pneumococcal pulmonary inflammation is required for high-level bacteremia and systemic infection, partly by disrupting lung epithelium through 12-LOX-dependent HXA3 production and subsequent PMN transepithelial migration.
Collapse
Affiliation(s)
- Rudra Bhowmick
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA 02111, USA
| | - Nang Maung
- Immune Disease Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Bryan P Hurley
- Mucosal Immunology Laboratory, Massachusetts General Hospital/Harvard Medical School, Charlestown, MA 02129, USA
| | - Elsa Bou Ghanem
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA 02111, USA
| | - Karsten Gronert
- Vision Science Program, School of Optometry, University of California, Berkeley, CA 94720, USA
| | - Beth A McCormick
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - John M Leong
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA 02111, USA
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Infection with Streptococcus pneumoniae (pneumococcus) results in colonization, which can lead to local or invasive disease, of which pneumonia is the most common manifestation. Despite the availability of pneumococcal vaccines, pneumococcal pneumonia is the leading cause of community and inhospital pneumonia in the United States and globally. This article discusses new insights into the pathogenesis of pneumococcal disease. RECENT FINDINGS The host-microbe interactions that determine whether pneumococcal colonization will result in clearance or invasive disease are highly complex. This article focuses on new information in three areas that bear on the pathogenesis of pneumococcal disease: factors that govern colonization, the prelude to invasive disease, including effects on colonization and invasion of capsular serotype, pneumolysin, surface proteins that regulate complement deposition, biofilm formation and agglutination; the effect of coinfection with other bacteria and viruses on pneumococcal growth and virulence, including the synergistic effect of influenza virus; and the contribution of the host inflammatory response to the pathogenesis of pneumococcal pneumonia, including the effects of pattern recognition molecules, cells that enhance and modulate inflammation, and therapies that modulate inflammation, such as statins. SUMMARY Recent research on pneumococcal pathogenesis reveals new mechanisms by which microbial factors govern the ability of pneumococcus to progress from the state of colonization to disease and host inflammatory responses contribute to the development of pneumonia. These mechanisms suggest that therapies which modulate the inflammatory response could hold promise for ameliorating damage stemming from the host inflammatory response in pneumococcal disease.
Collapse
|
26
|
Henriques-Normark B, Tuomanen EI. The pneumococcus: epidemiology, microbiology, and pathogenesis. Cold Spring Harb Perspect Med 2013; 3:3/7/a010215. [PMID: 23818515 DOI: 10.1101/cshperspect.a010215] [Citation(s) in RCA: 280] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The pneumococcus is the classic Gram-positive extracellular pathogen. The medical burden of diseases it causes is amongst the greatest in the world. Intense study for more than 100 years has yielded an understanding of fundamental aspects of its physiology, pathogenesis, and immunity. Efforts to control infection have led to the deployment of polysaccharide vaccines and an understanding of antibiotic resistance. The inflammatory response to pneumococci, one of the most potent in medicine, has revealed the double-edged sword of clearance of infection but at a cost of damage to host cells. In virtually every aspect of the infectious process, the pneumococcus has set the rules of the Gram-positive pathogenesis game.
Collapse
|
27
|
Interleukin-23 (IL-23) deficiency disrupts Th17 and Th1-related defenses against Streptococcus pneumoniae infection. Cytokine 2013; 64:375-81. [PMID: 23752068 DOI: 10.1016/j.cyto.2013.05.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 04/11/2013] [Accepted: 05/17/2013] [Indexed: 12/11/2022]
Abstract
Resolution of acute of infection caused by capsular Streptococcus pneumoniae infection in the absence of effective antibiotic therapy requires tight regulation of immune and inflammatory responses. To provide new mechanistic insight of the requirements needed for innate host defenses against acute S. pneumoniae infection, we examined how IL-23 deficiency mediated acute pulmonary resistance. We found that IL-23 deficient mice were more susceptible to bacterial colonization in the lungs corresponding with greater bacterial dissemination. The lack of IL-23 was found to decrease IL-6 and IL-12p70 cytokine levels in bronchiolar lavage within the initial day after infection. Pulmonary leukocytes isolated from infected IL-23 deficient mice demonstrated a dramatic decrease in IL-17A and IFN-γ in response to heat-killed organisms. These findings corresponded with significant abrogation of neutrophilic infiltrate in the lungs compared to IL-23 competent mice. Whereas previous studies have shown opposing influences of IL-12/IL-23 regulation, our findings suggest a concordant dependency of IL-23 expression on Th1 and Th17-related responses.
Collapse
|
28
|
Barquero-Calvo E, Martirosyan A, Ordoñez-Rueda D, Arce-Gorvel V, Alfaro-Alarcón A, Lepidi H, Malissen B, Malissen M, Gorvel JP, Moreno E. Neutrophils exert a suppressive effect on Th1 responses to intracellular pathogen Brucella abortus. PLoS Pathog 2013; 9:e1003167. [PMID: 23458832 PMCID: PMC3573106 DOI: 10.1371/journal.ppat.1003167] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 12/17/2012] [Indexed: 12/31/2022] Open
Abstract
Polymorphonuclear neutrophils (PMNs) are the first line of defense against microbial pathogens. In addition to their role in innate immunity, PMNs may also regulate events related to adaptive immunity. To investigate the influence of PMNs in the immune response during chronic bacterial infections, we explored the course of brucellosis in antibody PMN-depleted C57BL/6 mice and in neutropenic mutant Genista mouse model. We demonstrate that at later times of infection, Brucella abortus is killed more efficiently in the absence of PMNs than in their presence. The higher bacterial removal was concomitant to the: i) comparatively reduced spleen swelling; ii) augmented infiltration of epithelioid histiocytes corresponding to macrophages/dendritic cells (DCs); iii) higher recruitment of monocytes and monocyte/DCs phenotype; iv) significant activation of B and T lymphocytes, and v) increased levels of INF-γ and negligible levels of IL4 indicating a balance of Th1 over Th2 response. These results reveal that PMNs have an unexpected influence in dampening the immune response against intracellular Brucella infection and strengthen the notion that PMNs actively participate in regulatory circuits shaping both innate and adaptive immunity.
Collapse
Affiliation(s)
- Elías Barquero-Calvo
- Programa de Investigación en Enfermedades Tropicales, Escuela de Medicina Veterinaria, Universidad Nacional, Heredia, Costa Rica
| | - Anna Martirosyan
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
| | - Diana Ordoñez-Rueda
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
| | - Vilma Arce-Gorvel
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
| | - Alejandro Alfaro-Alarcón
- Departamento de Patología, Escuela de Medicina Veterinaria, Universidad Nacional, Heredia, Costa Rica
| | - Hubert Lepidi
- Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes (URMITE), Marseille, France
- Centre National de la Recherche Scientifique, (CNRS), UMR7278, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1095, Marseille, France
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
| | - Marie Malissen
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
| | - Jean-Pierre Gorvel
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, Marseille, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Marseille, France
- Centre National de la Recherche Scientifique (CNRS), UMR7280, Marseille, France
| | - Edgardo Moreno
- Programa de Investigación en Enfermedades Tropicales, Escuela de Medicina Veterinaria, Universidad Nacional, Heredia, Costa Rica
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| |
Collapse
|
29
|
Marriott HM, Daigneault M, Thompson AAR, Walmsley SR, Gill SK, Witcher DR, Wroblewski VJ, Hellewell PG, Whyte MKB, Dockrell DH. A decoy receptor 3 analogue reduces localised defects in phagocyte function in pneumococcal pneumonia. Thorax 2012; 67:985-92. [PMID: 22735687 PMCID: PMC3505869 DOI: 10.1136/thoraxjnl-2012-201591] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background Therapeutic strategies to modulate the host response to bacterial pneumonia are needed to improve outcomes during community-acquired pneumonia. This study used mice with impaired Fas signalling to examine susceptibility to pneumococcal pneumonia and decoy receptor 3 analogue (DcR3-a) to correct factors associated with increased susceptibility. Methods Wild-type mice and those with varying degrees of impairment of Fas (lpr) or Fas ligand signalling (gld) were challenged with Streptococcus pneumoniae and microbiological and immunological outcomes measured in the presence or absence of DcR3-a. Results During established pneumonia, neutrophils became the predominant cell in the airway and gld mice were less able to clear bacteria from the lungs, demonstrating localised impairment of pulmonary neutrophil function in comparison to lpr or wild-type mice. T-cells from gld mice had enhanced activation and reduced apoptosis in comparison to wild-type and lpr mice during established pneumonia. Treatment with DcR3-a reduced T-cell activation and corrected the defect in pulmonary bacterial clearance in gld mice. Conclusions The results suggest that imbalance in tumour necrosis factor superfamily signalling and excessive T-cell activation can impair bacterial clearance in the lung but that DcR3-a treatment can reduce T-cell activation, restore optimal pulmonary neutrophil function and enhance bacterial clearance during S pneumoniae infection.
Collapse
Affiliation(s)
- Helen M Marriott
- Department of Infection and Immunity, University of Sheffield, Sheffield, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Boyd AR, Hinojosa CA, Rodriguez PJ, Orihuela CJ. Impact of oral simvastatin therapy on acute lung injury in mice during pneumococcal pneumonia. BMC Microbiol 2012; 12:73. [PMID: 22587610 PMCID: PMC3438118 DOI: 10.1186/1471-2180-12-73] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 05/03/2012] [Indexed: 12/26/2022] Open
Abstract
Background Recent studies suggest that the reported protective effects of statins (HMG-CoA reductase inhibitors) against community-acquired pneumonia (CAP) and sepsis in humans may be due to confounders and a healthy user-effect. To directly test whether statins are protective against Streptococcus pneumoniae, the leading cause of CAP, we examined the impact of prolonged oral simvastatin therapy at physiologically relevant doses in a mouse model of pneumococcal pneumonia. BALB/c mice were placed on rodent chow containing 0 mg/kg (control), 12 mg/kg (low simvastatin diet [LSD]; corresponds to 1.0 mg/kg/day), or 120 mg/kg (high simvastatin diet [HSD]; corresponds to 10 mg/kg/day) simvastatin for four weeks, infected intratracheally with S. pneumoniae serotype 4 strain TIGR4, and sacrificed at 24, 36, or 42 h post-infection for assessment of lung histology, cytokine production, vascular leakage and edema, bacterial burden and bloodstream dissemination. Some mice received ampicillin at 12-h intervals beginning at 48 h post-infection and were monitored for survival. Immunoblots of homogenized lung samples was used to assess ICAM-1 production. Results Mice receiving HSD had reduced lung consolidation characterized by less macrophage and neutrophil infiltration and a significant reduction in the chemokines MCP-1 (P = 0.03) and KC (P = 0.02) and ICAM-1 in the lungs compared to control mice. HSD mice also had significantly lower bacterial titers in the blood at 36 (P = 0.007) and 42 (P = 0.03) hours post-infection versus controls. LSD had a more modest effect against S. pneumoniae but also resulted in reduced bacterial titers in the lungs and blood of mice after 42 h and a reduced number of infiltrated neutrophils. Neither LSD nor HSD mice had reduced mortality in a pneumonia model where mice received ampicillin 48 h after challenge. Conclusions Prolonged oral simvastatin therapy had a strong dose-dependent effect on protection against S. pneumoniae as evidenced by reduced neutrophil infiltration, maintenance of vascular integrity, and lowered chemokine production in the lungs of mice on HSD. Statin therapy also protected through reduced bacterial burden in the lungs. Despite these protective correlates, mortality in the simvastatin-receiving cohorts was equivalent to controls. Thus, oral simvastatin at physiologically relevant doses only modestly protects against pneumococcal pneumonia.
Collapse
Affiliation(s)
- Angela R Boyd
- Department of Microbiology and Immunology, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, USA
| | | | | | | |
Collapse
|
31
|
Neill DR, Fernandes VE, Wisby L, Haynes AR, Ferreira DM, Laher A, Strickland N, Gordon SB, Denny P, Kadioglu A, Andrew PW. T regulatory cells control susceptibility to invasive pneumococcal pneumonia in mice. PLoS Pathog 2012; 8:e1002660. [PMID: 22563306 PMCID: PMC3334885 DOI: 10.1371/journal.ppat.1002660] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 03/05/2012] [Indexed: 11/18/2022] Open
Abstract
Streptococcus pneumoniae is an important human pathogen responsible for a spectrum of diseases including pneumonia. Immunological and pro-inflammatory processes induced in the lung during pneumococcal infection are well documented, but little is known about the role played by immunoregulatory cells and cytokines in the control of such responses. We demonstrate considerable differences in the immunomodulatory cytokine transforming growth factor (TGF)-β between the pneumococcal pneumonia resistant BALB/c and susceptible CBA/Ca mouse strains. Immunohistochemistry and flow cytometry reveal higher levels of TGF-β protein in BALB/c lungs during pneumococcal pneumonia that correlates with a rapid rise in lung Foxp3+Helios+ T regulatory cells. These cells have protective functions during pneumococcal pneumonia, because blocking their induction with an inhibitor of TGF-β impairs BALB/c resistance to infection and aids bacterial dissemination from lungs. Conversely, adoptive transfer of T regulatory cells to CBA/Ca mice, prior to infection, prolongs survival and decreases bacterial dissemination from lungs to blood. Importantly, strong T regulatory cell responses also correlate with disease-resistance in outbred MF1 mice, confirming the importance of immunoregulatory cells in controlling protective responses to the pneumococcus. This study provides exciting new evidence for the importance of immunomodulation during pulmonary pneumococcal infection and suggests that TGF-β signalling is a potential target for immunotherapy or drug design. Streptococcus pneumoniae is a major human bacterial pathogen that causes a wide range of diseases including pneumonia, meningitis, sepsis and ear infections. The bacterium is responsible for around 1.2 million deaths per year, mostly in high-risk groups such as children, the elderly and those with a weakened immune system. Infection with the pneumococcus can induce a wide-variety of immune responses and disease symptoms and it is not known why some people are more resistant to infection than others. Here, we identify an important role in natural resistance against pneumococcal pneumonia for a group of cells – known as T regulatory cells – that control the immune response to pneumococcal infection. In mice, strong T regulatory cell responses correlate with resistance to invasive pneumococcal pneumonia. Disease-resistance can be boosted by administering T regulatory cells to highly susceptible mice or inhibited by blocking the activity of these cells in resistant mice. These results advance our understanding of the host immunity differences that underpin resistance to pneumococcal pneumonia and offer hope that in the future we might boost resistance in susceptible individuals through modulation of their immune system.
Collapse
Affiliation(s)
- Daniel R. Neill
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Vitor E. Fernandes
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Laura Wisby
- MRC Harwell, Mammalian Genetics Unit, Harwell Science and Innovation Campus, Oxfordshire, United Kingdom
| | - Andrew R. Haynes
- MRC Harwell, Mammalian Genetics Unit, Harwell Science and Innovation Campus, Oxfordshire, United Kingdom
| | - Daniela M. Ferreira
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Ameera Laher
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Natalie Strickland
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Stephen B. Gordon
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Paul Denny
- MRC Harwell, Mammalian Genetics Unit, Harwell Science and Innovation Campus, Oxfordshire, United Kingdom
| | - Aras Kadioglu
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
- * E-mail: (AK); (PWA)
| | - Peter W. Andrew
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
- * E-mail: (AK); (PWA)
| |
Collapse
|
32
|
A serotype 3 pneumococcal capsular polysaccharide-specific monoclonal antibody requires Fcγ receptor III and macrophages to mediate protection against pneumococcal pneumonia in mice. Infect Immun 2012; 80:1314-22. [PMID: 22290146 DOI: 10.1128/iai.06081-11] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Antibodies to pneumococcal capsular polysaccharide (PPS) are required for PPS-based vaccine-mediated protection against Streptococcus pneumoniae. Previous work established that 1E2, a mouse IgG1 to PPS3 that does not induce serotype 3 (ST3) S. pneumoniae killing by phagocytes in vitro, protects mice from death after intranasal infection with ST3, but its efficacy was abrogated in FcγR (F common gamma receptor)-deficient mice. In this study, we determined whether 1E2 efficacy against pulmonary ST3 infection requires FcγRIII. 1E2 did not protect FcγRIII-deficient (FcγRIII(-/-)) mice. Studies of the mechanism of 1E2-mediated effects showed that it resulted in a marked reduction in lung inflammation in ST3-infected wild-type (Wt [C57BL/6]) mice that was abrogated in FcγRIII(-/-) mice. 1E2 had no effect on early bacterial clearance in the lungs of ST3-infected Wt, FcγRIIB(-/-), or FcγRIII(-/-) mice, but it reduced levels of bacteremia and serum macrophage inflammatory protein-2) (MIP-2), interleukin-6 (IL-6), and tumor necrosis factor alpha (TNF-α) in Wt and FcγRIIB(-/-) mice, strains in which it is protective. As previous work showed that neutrophils were dispensable for 1E2 efficacy, we investigated whether macrophages are required for 1E2 efficacy against intranasal infection with ST3 and found that its efficacy was abrogated in Wt mice depleted of macrophages intranasally. In vitro studies revealed that1E2 promoted ST3 internalization by naïve alveolar macrophages but did not induce early intracellular killing. Macrophages from 1E2-treated ST3-infected mice studied ex vivo exhibited more apoptosis than those from FcγRIII(-/-) mice. These findings suggest that 1E2 mediates protection against ST3 in mice by affecting the inflammatory response, perhaps in part via macrophage apoptosis, rather than by inducing early bacterial clearance.
Collapse
|
33
|
Interleukin-1β regulates CXCL8 release and influences disease outcome in response to Streptococcus pneumoniae, defining intercellular cooperation between pulmonary epithelial cells and macrophages. Infect Immun 2011; 80:1140-9. [PMID: 22158745 DOI: 10.1128/iai.05697-11] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The success of Streptococcus pneumoniae (the pneumococcus) as a pulmonary pathogen is related to its restriction of innate immune responses by respiratory epithelial cells. The mechanisms used to overcome this restriction are incompletely elucidated. Pulmonary chemokine expression involves complex cellular and molecular networks, involving the pulmonary epithelium, but the specific cellular interactions and the cytokines that control them are incompletely defined. We show that serotype 2 or 4 pneumococci induce only modest levels of CXCL8 expression from epithelial cell lines, even in the absence of a polysaccharide capsule. In contrast, coculture of A549 cells with the macrophage-like THP-1 cell line, differentiated with vitamin D, or monocyte-derived macrophages enhanced CXCL8 release. Supernatants from the THP-1 cell line prime A549 cells to release CXCL8 at levels similar to cocultures. Interleukin-1Ra (IL-1Ra) inhibits CXCL8 release from cocultures and reduces the activity of macrophage-conditioned media, but inhibition of tumor necrosis factor alpha (TNF-α) had only a minimal effect on CXCL8 release. Release of IL-1β but not TNF-α was upregulated in cocultures. IL-1 type 1 receptor knockout C57BL/6 and BALB/c mice confirmed the importance of IL-1 signaling in CXC chemokine expression and neutrophil recruitment in vivo. In fulminant disease, increased IL-1 signaling resulted in increased neutrophils in the airway and more invasive disease. These results demonstrate that IL-1 is an important component of the cellular network involving macrophages and epithelial cells, which facilitates CXC chemokine expression and aids neutrophil recruitment during pneumococcal pneumonia. They also highlight a potential clinical role for anti-IL-1 treatment to limit excessive neutrophilic inflammation in the lung.
Collapse
|
34
|
Kadioglu A, Cuppone AM, Trappetti C, List T, Spreafico A, Pozzi G, Andrew PW, Oggioni MR. Sex-based differences in susceptibility to respiratory and systemic pneumococcal disease in mice. J Infect Dis 2011; 204:1971-9. [PMID: 22021621 DOI: 10.1093/infdis/jir657] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Systemic infection with Streptococcus pneumoniae was investigated in male and female mice in models of invasive pneumonia and sepsis. Male mice were found to be more susceptible to infection, exhibiting greater weight loss, marked decrease in body temperature, and a significantly higher mortality rate compared with female mice. For pneumonia, there were significant differences in survival rates. Female mice cleared their lung infections over time, whereas male mice, compared with female mice, had significantly increased numbers of colony-forming units in early stages of infection accompanied by higher levels of neutrophil recruitment in the first 24 hours after infection. Importantly, there were significant increases in proinflammatory cytokine levels during both sepsis and pneumonia in male compared with female mice. These cytokines were indicative of T-helper 1-type responses. The data presented here describe surprising differences in survival rates, neutrophil recruitment, and proinflammatory cytokine levels, indicating a sex-based difference in susceptibility to respiratory and systemic pneumococcal disease.
Collapse
Affiliation(s)
- Aras Kadioglu
- Department of Infection, Immunity, and Inflammation, University of Leicester, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Banerjee A, Kim BJ, Carmona EM, Cutting AS, Gurney MA, Carlos C, Feuer R, Prasadarao NV, Doran KS. Bacterial Pili exploit integrin machinery to promote immune activation and efficient blood-brain barrier penetration. Nat Commun 2011; 2:462. [PMID: 21897373 PMCID: PMC3195231 DOI: 10.1038/ncomms1474] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 08/10/2011] [Indexed: 12/25/2022] Open
Abstract
Group B Streptococcus (GBS) is the leading cause of meningitis in newborn infants. Bacterial cell surface appendages, known as pili, have been recently described in streptococcal pathogens, including GBS. The pilus tip adhesin, PilA, contributes to GBS adherence to blood-brain barrier (BBB) endothelium; however, the host receptor and the contribution of PilA in central nervous system (CNS) disease pathogenesis are unknown. Here we show that PilA binds collagen, which promotes GBS interaction with the α₂β₁ integrin resulting in activation of host chemokine expression and neutrophil recruitment during infection. Mice infected with the PilA-deficient mutant exhibit delayed mortality, a decrease in neutrophil infiltration and bacterial CNS dissemination. We find that PilA-mediated virulence is dependent on neutrophil influx as neutrophil depletion results in a decrease in BBB permeability and GBS-BBB penetration. Our results suggest that the bacterial pilus, specifically the PilA adhesin, has a dual role in immune activation and bacterial entry into the CNS.
Collapse
Affiliation(s)
- Anirban Banerjee
- Department of Biology and Center for Microbial Sciences, San Diego State University, 5500 Campanile Drive, San Diego, California 92182, USA
| | - Brandon J. Kim
- Department of Biology and Center for Microbial Sciences, San Diego State University, 5500 Campanile Drive, San Diego, California 92182, USA
- These authors contributed equally to this work
| | - Ellese M. Carmona
- Department of Biology and Center for Microbial Sciences, San Diego State University, 5500 Campanile Drive, San Diego, California 92182, USA
- These authors contributed equally to this work
| | - Andrew S. Cutting
- Department of Biology and Center for Microbial Sciences, San Diego State University, 5500 Campanile Drive, San Diego, California 92182, USA
| | - Michael A. Gurney
- Department of Biology and Center for Microbial Sciences, San Diego State University, 5500 Campanile Drive, San Diego, California 92182, USA
| | - Chris Carlos
- Department of Biology and Center for Microbial Sciences, San Diego State University, 5500 Campanile Drive, San Diego, California 92182, USA
| | - Ralph Feuer
- Department of Biology and Center for Microbial Sciences, San Diego State University, 5500 Campanile Drive, San Diego, California 92182, USA
| | - Nemani V. Prasadarao
- Division of Infectious Diseases, Department of Pediatrics, Childrens Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles, California 90027, USA
| | - Kelly S. Doran
- Department of Biology and Center for Microbial Sciences, San Diego State University, 5500 Campanile Drive, San Diego, California 92182, USA
- Department of Pediatrics, University of California–San Diego School of Medicine, La Jolla, California 92093, USA
| |
Collapse
|
36
|
The innate immune response to Streptococcus pneumoniae in the lung depends on serotype and host response. Vaccine 2011; 29:8002-11. [PMID: 21864623 DOI: 10.1016/j.vaccine.2011.08.064] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Revised: 07/22/2011] [Accepted: 08/12/2011] [Indexed: 11/24/2022]
Abstract
Bacteremic pneumonia with some pneumococcal capsular serotypes, including serotype 3 (ST3), has been associated with a higher risk of death, whereas others, such as ST8, are associated with a lower risk. To provide a molecular basis for understanding such differences, we used oligo cDNA microarrays to analyze and compare the gene expression profiles of the lungs of Balb/c mice infected intranasally with either ST3, strain A66.1, or ST8, strain ATCC 6308 (6308). Compared to uninfected controls, infection with either A66.1 or 6308 led to inoculum-dependent expression of IFN-γ inducible CXC chemokines among other pro-inflammatory genes. To investigate the role that IFN-γ inducible chemokines CXCL9, CXCL10 and CXCL11 play in A66.1- and 6308-induced pneumonia, we examined the effect of the absence of their common receptor, CXCR3, on intranasal infection in CXCR3(-/-) (Balb/c) mice. Compared to wild type (WT) mice, virulence of A66.1 but not 6308 was attenuated in CXCR3(-/-) mice. A66.1-infected CXCR3(-/-) mice had fewer lung neutrophils and more alveolar macrophages 48 h after infection and fewer blood CFU 72 h after infection. Histopathological examination of lung sections revealed less inflammation among A66.1-infected CXCR3(-/-) than WT mice. The reduced virulence of A66.1 in CXCR3(-/-) mice suggests that inhibition of the functional activity of IFN-γ inducible chemokines modulates the host response to A66.1, in turn suggesting a novel approach to improve vaccine-mediated protection against ST3 pneumonia.
Collapse
|
37
|
Zhao L, KuoLee R, Harris G, Tram K, Yan H, Chen W. c-di-GMP protects against intranasal Acinetobacter baumannii infection in mice by chemokine induction and enhanced neutrophil recruitment. Int Immunopharmacol 2011; 11:1378-83. [PMID: 21496497 DOI: 10.1016/j.intimp.2011.03.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Revised: 03/28/2011] [Accepted: 03/31/2011] [Indexed: 12/25/2022]
Abstract
Acinetobacter baumannii has emerged as a major cause of both community-associated and nosocomial infections worldwide. A. baumannii rapidly develops resistance to multiple antibiotics; as a result, infections by this pathogen have become increasingly difficult to treat. In this study, we evaluated the effect of 3',5'-cyclic diguanylic acid (c-di-GMP), a bacterial second messenger and immunomodulator, in the host defense against A. baumannii infection in a mouse model of intranasal infection. Our results showed that 50 μg of c-di-GMP administered 18 h prior to infection provided the best protection against intranasal infection with A. baumannii. Mechanistically, administration of c-di-GMP induced the production of chemokines KC, MCP-1, MIP-1α, MIP-2 and RANTES, and enhanced neutrophil recruitment in the lung. Moreover, depletion of neutrophils abolished the protective role of c-di-GMP. Taken together, our data suggest that c-di-GMP confers resistance against intranasal A. baumannii infection in mice through a neutrophil-dependent mechanism and that c-di-GMP should be further explored as an immunomodulator for the treatment of A. baumannii infection.
Collapse
Affiliation(s)
- Lisa Zhao
- Institute for Biological Sciences, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario, Canada K1A 0R6
| | | | | | | | | | | |
Collapse
|
38
|
Coleman JR, Papamichail D, Yano M, García-Suárez MDM, Pirofski LA. Designed reduction of Streptococcus pneumoniae pathogenicity via synthetic changes in virulence factor codon-pair bias. J Infect Dis 2011; 203:1264-73. [PMID: 21343143 DOI: 10.1093/infdis/jir010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In this study, we used a previously described method of controlling gene expression with computer-based gene design and de novo DNA synthesis to attenuate the virulence of Streptococcus pneumoniae. We produced 2 S. pneumoniae serotype 3 (SP3) strains in which the pneumolysin gene (ply) was recoded with underrepresented codon pairs while retaining its amino acid sequence and determined their ply expression and pneumolysin production in vitro and their virulence in a mouse pulmonary infection model. Expression of ply and production of pneumolysin of the recoded SP3 strains were decreased, and the recoded SP3 strains were less virulent in mice than the wild-type SP3 strain or a Δply SP3 strain. Further studies showed that the least virulent recoded strain induced a markedly reduced inflammatory response in the lungs compared with the wild-type or Δply strain. These findings suggest that reducing pneumococcal virulence gene expression by altering codon-pair bias could hold promise for rational design of live-attenuated pneumococcal vaccines.
Collapse
Affiliation(s)
- J Robert Coleman
- Department of Medicine, Division of Infectious Disease, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, USA
| | | | | | | | | |
Collapse
|
39
|
Bewley MA, Marriott HM, Tulone C, Francis SE, Mitchell TJ, Read RC, Chain B, Kroemer G, Whyte MKB, Dockrell DH. A cardinal role for cathepsin d in co-ordinating the host-mediated apoptosis of macrophages and killing of pneumococci. PLoS Pathog 2011; 7:e1001262. [PMID: 21298030 PMCID: PMC3029254 DOI: 10.1371/journal.ppat.1001262] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Accepted: 12/21/2010] [Indexed: 11/23/2022] Open
Abstract
The bactericidal function of macrophages against pneumococci is enhanced by their apoptotic demise, which is controlled by the anti-apoptotic protein Mcl-1. Here, we show that lysosomal membrane permeabilization (LMP) and cytosolic translocation of activated cathepsin D occur prior to activation of a mitochondrial pathway of macrophage apoptosis. Pharmacological inhibition or knockout of cathepsin D during pneumococcal infection blocked macrophage apoptosis. As a result of cathepsin D activation, Mcl-1 interacted with its ubiquitin ligase Mule and expression declined. Inhibition of cathepsin D had no effect on early bacterial killing but inhibited the late phase of apoptosis-associated killing of pneumococci in vitro. Mice bearing a cathepsin D−/− hematopoietic system demonstrated reduced macrophage apoptosis in vivo, with decreased clearance of pneumococci and enhanced recruitment of neutrophils to control pulmonary infection. These findings establish an unexpected role for a cathepsin D-mediated lysosomal pathway of apoptosis in pulmonary host defense and underscore the importance of apoptosis-associated microbial killing to macrophage function. Tissue macrophages frequently undergo a program of cell death, termed apoptosis, following sustained ingestion and killing of bacteria. In macrophages, induction of apoptosis enhances bacterial killing when macrophages' initial killing capacity is exhausted. We have investigated the mechanism of apoptosis in macrophages exposed to pneumococci, the commonest cause of bacterial pneumonia. We show that the cell structure containing ingested bacteria, the phagolysosome, becomes permeabilized early in the death process. Pneumococcal exposure activates a phagolysosomal enzyme, cathepsin D, which induces apoptosis. Cathepsin D activation is required for permeabilization of mitochondria, an organelle implicated in apoptosis induction. Cathepsin D reduces levels of a negative regulator of apoptosis in macrophages, Mcl-1, by enhancing its association with an enzyme, which mediates its degradation. The importance of these findings was confirmed in a bone marrow transplant model in which mice either received bone marrow from mice containing or lacking the cathepsin D gene. This model showed that reduced apoptosis of alveolar macrophages occurred when cathepsin D was lacking, and that this impaired clearance of pneumococci in the mouse lung. We conclude that during bacterial challenge, lysosomal permeabilization and cathepsin D activation triggers a novel death pathway, in a timely fashion, linking bacterial killing to apoptosis induction.
Collapse
Affiliation(s)
- Martin A Bewley
- Medical School, University of Sheffield, Sheffield, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
KuoLee R, Harris G, Conlan JW, Chen W. Role of neutrophils and NADPH phagocyte oxidase in host defense against respiratory infection with virulent Francisella tularensis in mice. Microbes Infect 2011; 13:447-56. [PMID: 21277990 DOI: 10.1016/j.micinf.2011.01.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Revised: 01/11/2011] [Accepted: 01/14/2011] [Indexed: 02/04/2023]
Abstract
Francisella tularensis subspecies (subsp.) tularensis is a CDC Category A biological warfare agent and inhalation of as few as 15 bacilli can initiate severe disease. Relatively little is known about the cellular and molecular mechanisms of host defense against respiratory infection with subsp. tularensis. In this study, we examined the role of neutrophils and NADPH phagocyte oxidase in host resistance to pulmonary infection in a mouse intranasal infection model. We found that despite neutrophil recruitment to the lungs and increased concentrations of neutrophil-chemotactic chemokines (KC, MIP-2 and RANTES) in the bronchoalveolar lavage fluid following intranasal inoculation of the pathogen, neither depletion of neutrophils nor enhancement of their recruitment into the lungs had any impact on bacterial burdens or survival rate/time. Nevertheless, mice deficient in NADPH phagocyte oxidase (gp91(phox⁻/⁻)) did exhibit higher tissue and blood bacterial burdens and succumbed to infection one day earlier than wild-type C57BL/6 mice. These results imply that although neutrophils are not a major effector cell in defense against subsp. tularensis infection, NADPH phagocyte oxidase does play a marginal role.
Collapse
Affiliation(s)
- Rhonda KuoLee
- Institute for Biological Sciences, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario K1A0R6, Canada
| | | | | | | |
Collapse
|
41
|
Weber SE, Tian H, Pirofski LA. CD8+ cells enhance resistance to pulmonary serotype 3 Streptococcus pneumoniae infection in mice. THE JOURNAL OF IMMUNOLOGY 2010; 186:432-42. [PMID: 21135172 DOI: 10.4049/jimmunol.1001963] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Despite the success of the pneumococcal conjugate vaccine, pneumococcal pneumonia remains a significant clinical problem, and there is still much to learn about natural resistance and cellular immunity to pneumococcus. We investigated the role of T lymphocytes in resistance to serotype (ST) 3 Streptococcus pneumoniae in an intranasal infection model in C57BL/6 (wild-type [Wt]) and CD8(+) (CD8(-/-))- and CD4(+) (MHC class II(-/-))-deficient mice. CD8(-/-) mice exhibited significantly more bacterial dissemination and lung inflammation and a significantly more lethal phenotype than Wt mice. However, there was no difference in the bacterial dissemination, lung inflammation, or survival of Wt and MHC class II(-/-) mice. Perforin (Pfn)(-/-) and IFN-γ(-/-) mice, which were used to dissect the role of CD8(+) T cells in our model, also exhibited a more lethal survival phenotype than Wt mice. Comparison of lung chemokine/cytokine levels by Luminex and cellular recruitment by FACS in Wt mice and knockout strains revealed that CD8(-/-) and IFN-γ(-/-) mice, which had the most lethal survival phenotype, had more CD4(+)IL-17(+) T (Th17) cells, IL-17, neutrophil chemoattractants, and lung neutrophils, and fewer regulatory T cells than Wt mice. CD4(+) T cell depletion improved the survival of ST-infected CD8(-/-) mice, and survival studies in Th17-deficient mice revealed that the Th17 response was dispensable for ST3 resistance in our model. Taken together, these findings demonstrate that CD8(+) cells are required, but CD4(+) T cells are dispensable for resistance to ST3 pneumonia in mice and suggest a previously unsuspected role for CD8(+) cells in modulating the inflammatory response to ST3.
Collapse
Affiliation(s)
- Sarah E Weber
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | |
Collapse
|
42
|
Ocuin LM, Bamboat ZM, Balachandran VP, Cavnar MJ, Obaid H, Plitas G, DeMatteo RP. Neutrophil IL-10 suppresses peritoneal inflammatory monocytes during polymicrobial sepsis. J Leukoc Biol 2010; 89:423-32. [PMID: 21106642 DOI: 10.1189/jlb.0810479] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Septic peritonitis remains a major cause of death. Neutrophils and inflammatory monocytes are principal components of the innate immune system and are essential for defense against a range of microbial pathogens. Their role and interaction in polymicrobial sepsis have not been defined clearly. Using a murine model of CLP to induce moderate sepsis, we found that neutrophil depletion did not alter survival, whereas depletion of neutrophils and inflammatory monocytes markedly reduced survival. After neutrophil depletion, inflammatory monocytes had greater phagocytic capacity and oxidative burst, and increased expression of costimulatory molecules, TNF, and iNOS. Notably, peritoneal neutrophils produced IL-10 following CLP. Adoptive i.p. transfer of WT but not IL-10(-/-) neutrophils into septic mice reduced monocyte expression of TNF. In vitro experiments confirmed that monocyte suppression was mediated by neutrophil-derived IL-10. Thus, during septic peritonitis, neutrophils suppress peritoneal inflammatory monocytes through IL-10 and are dispensable for survival.
Collapse
Affiliation(s)
- Lee M Ocuin
- Hepatopancreatobiliary Service, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Characterization of gene use and efficacy of mouse monoclonal antibodies to Streptococcus pneumoniae serotype 8. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 18:59-66. [PMID: 21068211 DOI: 10.1128/cvi.00368-10] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Streptococcus pneumoniae is the most common cause of community-acquired pneumonia in the United States and globally. Despite the availability of pneumococcal capsular polysaccharide (PPS) and protein conjugate-based vaccines, the prevalence of antibiotic-resistant pneumococcal strains, serotype (ST) replacement in nonconjugate vaccine strains, and uncertainty as to whether the PPS vaccine that is used in adults protects against pneumonia emphasize the need for continued efforts to understand the nature of protective PPS antibody responses. In this study, we generated mouse monoclonal antibodies (MAbs) to a conjugate consisting of the PPS of serotype 8 (PPS8) S. pneumoniae and tetanus toxoid. Thirteen MAbs, including four IgMs that bound to PPS8 and phosphorylcholine (PC) and five IgMs and four IgG1s that bound to PPS8 but not PC, were produced, and their nucleotide sequences, epitope and fine specificity, and efficacy against lethal challenge with ST8 S. pneumoniae were determined. MAbs that bound to PPS8 exhibited gene use that was distinct from that exhibited by MAbs that bound to PC. Only PPS8-binding MAbs that did not bind PC were protective in mice. All 13 MAbs used germ line variable-region heavy (V(H)) and light (V(L)) chain genes, with no evidence of somatic hypermutation. Our data reveal a relationship between PPS specificity and V(H) gene use and MAb efficacy in mice. These findings provide insight into the relationship between antibody molecular structure and function and hold promise for the development of novel surrogates for pneumococcal vaccine efficacy.
Collapse
|
44
|
Fabrizio K, Manix C, Tian H, van Rooijen N, Pirofski LA. The efficacy of pneumococcal capsular polysaccharide-specific antibodies to serotype 3 Streptococcus pneumoniae requires macrophages. Vaccine 2010; 28:7542-50. [PMID: 20800700 DOI: 10.1016/j.vaccine.2010.08.061] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Revised: 08/01/2010] [Accepted: 08/12/2010] [Indexed: 10/19/2022]
Abstract
The efficacy of antibody immunity against Streptococcus pneumoniae stems from the ability of opsonic, serotype (ST)-specific antibodies to pneumococcal capsular polysaccharide (PPS) to facilitate killing of the homologous ST by host phagocytes. However, PPS-specific antibodies have been identified that are protective in mice, but do not promote opsonic killing in vitro, raising the question of how they mediate protection in vivo. To probe this question, we investigated the dependence of antibody efficacy against lethal systemic (intraperitoneal, i.p.) infection with Streptococcus pneumoniae serotype 3 (ST3) on macrophages and neutrophils for the following PPS3-specific monoclonal antibodies (MAbs) in survival experiments in mice using a non-opsonic human IgM (A7), a non-opsonic mouse IgG1 (1E2) and an opsonic mouse IgG1 (5F6). The survival of A7- and PPS3-specific and isotype control MAb-treated neutrophil-depleted and neutrophil-sufficient and macrophage-depleted and macrophage-sufficient mice were determined after i.p. challenge with ST3 strains 6303 and WU2. Neutrophils were dispensable for A7 and the mouse MAbs to mediate protection in this model, but macrophages were required for the efficacy of A7 and optimal mouse MAb-mediated protection. For A7-treated mice, macrophage-depleted mice had higher blood CFU, cytokines and peripheral neutrophil levels than macrophage-sufficient mice, and macrophage-sufficient mice had lower tissue bacterial burdens than control MAb-treated mice. These findings demonstrate that macrophages contribute to opsonic and non-opsonic PPS3-specific MAb-mediated protection against ST3 infection by enhancing bacterial clearance and suggest that neutrophils do not compensate for the absence of macrophages in the model used in this study.
Collapse
Affiliation(s)
- Kevin Fabrizio
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | |
Collapse
|
45
|
Mucosal administration of flagellin protects mice from Streptococcus pneumoniae lung infection. Infect Immun 2010; 78:4226-33. [PMID: 20643849 DOI: 10.1128/iai.00224-10] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Streptococcus pneumoniae is a major cause of pneumonia in infants and the elderly. Innate defenses are essential to the control of pneumococcal infections, and deficient responses can trigger disease in susceptible individuals. Here we showed that flagellin can locally activate innate immunity and thereby increase the resistance to acute pneumonia. Flagellin mucosal treatment improved S. pneumoniae clearance in the lungs and promoted increased survival of infection. In addition, lung architecture was fully restored after the treatment of infected mice, indicating that flagellin allows the reestablishment of steady-state conditions. Using a flagellin mutant that is unable to signal through Toll-like receptor 5 (TLR5), we established that TLR5 signaling is essential for protection. In the respiratory tract, flagellin induced neutrophil infiltration into airways and upregulated the expression of genes coding for interleukin 6 (IL-6), tumor necrosis factor alpha (TNF-α), CXCL1, CXCL2, and CCL20. Using depleting antibodies, we demonstrated that neutrophils are major effectors of protection. Further, we found that B- and T-cell-deficient SCID mice clear S. pneumoniae challenge to the same extent as immunocompetent animals, suggesting that these cell populations are not required for flagellin-induced protection. In conclusion, this study emphasizes that mucosal stimulation of innate immunity by a TLR not naturally engaged by S. pneumoniae can increase the potential to cure pneumococcal pneumonia.
Collapse
|
46
|
Schabbauer G, Matt U, Günzl P, Warszawska J, Furtner T, Hainzl E, Elbau I, Mesteri I, Doninger B, Binder BR, Knapp S. Myeloid PTEN promotes inflammation but impairs bactericidal activities during murine pneumococcal pneumonia. THE JOURNAL OF IMMUNOLOGY 2010; 185:468-76. [PMID: 20505137 DOI: 10.4049/jimmunol.0902221] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Phosphatidylinositol 3-kinase has been described as an essential signaling component involved in the chemotactic cell influx that is required to eliminate pathogens. At the same time, PI3K was reported to modulate the immune response, thus limiting the magnitude of acute inflammation. The precise role of the PI3K pathway and its endogenous antagonist phosphatase and tensin homolog deleted on chromosome 10 (PTEN) during clinically relevant bacterial infections is still poorly understood. Utilizing mice lacking myeloid cell-specific PTEN, we studied the impact of PTEN on the immune response to Streptococcus pneumoniae. Survival analysis disclosed that PTEN-deficient mice displayed less severe signs of disease and prolonged survival. The inflammatory response to S. pneumoniae was greatly reduced in macrophages in vitro and in vivo. Unexpectedly, neutrophil influx to the lungs was significantly impaired in animals lacking myeloid-cell PTEN, whereas the additional observation of improved phagocytosis by alveolar macrophages lacking PTEN ultimately resulted in unaltered lung CFUs following bacterial infection. Together, the absence of myeloid cell-associated PTEN and consecutively enhanced PI3K activity dampened pulmonary inflammation, reduced neutrophil influx, and augmented phagocytic properties of macrophages, which ultimately resulted in decreased tissue injury and improved survival during murine pneumococcal pneumonia.
Collapse
Affiliation(s)
- Gernot Schabbauer
- Department of Vascular Biology and Thrombosis Research, Center for Biomolecular Medicine and Pharmacology, Austrian Academy of Sciences, Vienna, Austria.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Cruse G, Fernandes VE, de Salort J, Pankhania D, Marinas MS, Brewin H, Andrew PW, Bradding P, Kadioglu A. Human lung mast cells mediate pneumococcal cell death in response to activation by pneumolysin. THE JOURNAL OF IMMUNOLOGY 2010; 184:7108-15. [PMID: 20483738 DOI: 10.4049/jimmunol.0900802] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mast cells are emerging as contributors to innate immunity. Mouse mast cells have a pivotal role in protection against bacterial infection, and human cord blood-derived mast cells reduce bacterial viability in culture. The objectives of this study were to determine whether human lung mast cells (HLMCs) might be protective against pneumococcal lung infection through direct antimicrobial activity. Tissue-derived HLMCs and the human mast cell lines HMC-1 and LAD2 were cocultured with wild-type and mutant pneumococci, and viability and functional assays were performed. Mast cells were also stimulated with purified pneumolysin. HLMCs killed wild-type serotype-2 (D39) pneumococci in coculture but had no effect on an isogenic pneumolysin-deficient (PLN-A) pneumococcus. D39 wild-type, but not PLN-A pneumococci, induced the release of leukotriene C4 from human mast cells in a dose-dependent manner, which was not accompanied by histamine release. Stimulation of mast cells with sublytic concentrations of purified pneumolysin replicated this effect. Furthermore, pneumolysin induced the release of the cathelicidin LL-37 from HLMCs, purified LL-37 reduced pneumococcal viability, and neutralizing Ab to LL-37 attenuated mast cell-dependent pneumococcal killing. In addition, at high concentrations, all pneumococcal strains tested reduced HLMC viability through a combination of pneumolysin and H2O2-dependent mechanisms. HLMCs exhibit direct antimicrobial activity to pneumococci through their activation by pneumolysin. This antimicrobial activity is mediated, in part, by the release of LL-37 from HLMCs. This suggests that mast cells provide an early warning system and potentially limit pneumococcal dissemination early in the course of invasive pulmonary pneumococcal disease.
Collapse
Affiliation(s)
- Glenn Cruse
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester Medical School, Leicester, United Kingdom.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Subramaniam KS, Datta K, Quintero E, Manix C, Marks MS, Pirofski LA. The absence of serum IgM enhances the susceptibility of mice to pulmonary challenge with Cryptococcus neoformans. THE JOURNAL OF IMMUNOLOGY 2010; 184:5755-67. [PMID: 20404271 DOI: 10.4049/jimmunol.0901638] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The importance of T cell-mediated immunity for resistance to the disease (cryptococcal disease) caused by Cryptococcus neoformans is incontrovertible, but whether Ab immunity also contributes to resistance remains uncertain. To investigate the role of IgM in resistance to C. neoformans, we compared the survival, fungal burden, lung and brain inflammatory responses, and lung phagocytic response of sIgM(-/-) mice, which lack secreted IgM, to that of IgM sufficient C57BL6x129Sv (heretofore, control) mice at different times after intranasal infection with C. neoformans (24067). sIgM(-/-) mice had higher mortality and higher blood and brain CFUs 28 d postinfection, but lung CFUs were comparable. Lungs of control mice manifested exuberant histiocytic inflammation with visible C. neoformans, findings that were not observed in sIgM(-/-) mice, whereas in brain sections, sIgM(-/-) mice had marked inflammation with visible C. neoformans that was not observed in control mice. Cytokine responses were significant for higher levels of lung IL-1beta and IL-12 24 h postinfection in control mice and higher levels of lung and brain IL-17 28 d postinfection in sIgM(-/-) mice. Alveolar macrophage phagocytosis was significantly higher for control than for sIgM(-/-) mice 24 h postinfection; however, phagocytic indices of sIgM(-/-) mice increased after reconstitution of sIgM(-/-) mice with polyclonal IgM. These data establish a previously unrecognized role for IgM in resistance to intranasal infection with C. neoformans in mice and suggest that the mechanism by which it mediates a host benefit is by augmenting Th1 polarization, macrophage recruitment and phagocytosis of C. neoformans.
Collapse
Affiliation(s)
- Krishanthi S Subramaniam
- Department of Microbiology and Immunology, Albert Einstein College of Medicine of the Yeshiva University, Bronx, NY 10461, USA
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
Pneumococcus remains the most common cause of community-acquired pneumonia worldwide. Streptococcus pneumoniae is well adapted to people, and is a frequent inhabitant of the upper airways in healthy hosts. This seemingly innocuous state of colonisation is a dynamic and competitive process in which the pathogen attempts to engage the host, proliferate, and invade the lower airways. The host in turn continuously deploys an array of innate and acquired cellular and humoral defences to prevent pneumococci from breaching tissue barriers. Discoveries into essential molecular mechanisms used by pneumococci to evade host-sensing systems that are designed to contain the pathogen provide new insights into potential treatment options. Versatility of the genome of pneumococci and the bacteria's polygenic virulence capabilities show that a multifaceted approach with many vaccine antigens, antibiotic combinations, and immunoadjuvant therapies will be needed to control this microbe.
Collapse
Affiliation(s)
- Tom van der Poll
- Centre for Infection and Immunity Amsterdam, Centre for Experimental and Molecular Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, the Netherlands.
| | | |
Collapse
|
50
|
Koedel U, Frankenberg T, Kirschnek S, Obermaier B, Häcker H, Paul R, Häcker G. Apoptosis is essential for neutrophil functional shutdown and determines tissue damage in experimental pneumococcal meningitis. PLoS Pathog 2009; 5:e1000461. [PMID: 19478887 PMCID: PMC2682662 DOI: 10.1371/journal.ppat.1000461] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2008] [Accepted: 04/30/2009] [Indexed: 01/10/2023] Open
Abstract
During acute bacterial infections such as meningitis, neutrophils enter the tissue where they combat the infection before they undergo apoptosis and are taken up by macrophages. Neutrophils show pro-inflammatory activity and may contribute to tissue damage. In pneumococcal meningitis, neuronal damage despite adequate chemotherapy is a frequent clinical finding. This damage may be due to excessive neutrophil activity. We here show that transgenic expression of Bcl-2 in haematopoietic cells blocks the resolution of inflammation following antibiotic therapy in a mouse model of pneumococcal meningitis. The persistence of neutrophil brain infiltrates was accompanied by high levels of IL-1β and G-CSF as well as reduced levels of anti-inflammatory TGF-β. Significantly, Bcl-2-transgenic mice developed more severe disease that was dependent on neutrophils, characterized by pronounced vasogenic edema, vasculitis, brain haemorrhages and higher clinical scores. In vitro analysis of neutrophils demonstrated that apoptosis inhibition completely preserves neutrophil effector function and prevents internalization by macrophages. The inhibitor of cyclin-dependent kinases, roscovitine induced apoptosis in neutrophils in vitro and in vivo. In wild type mice treated with antibiotics, roscovitine significantly improved the resolution of the inflammation after pneumococcal infection and accelerated recovery. These results indicate that apoptosis is essential to turn off activated neutrophils and show that inflammatory activity and disease severity in a pyogenic infection can be modulated by targeting the apoptotic pathway in neutrophils. Infections are typically accompanied by inflammation and the presence of leucocytes at the infected site. Especially in pyogenic (pus generating) infections, neutrophils are the main leukocytes that arrive and that combat the infection. When the bacteria have been killed, the inflammation also has to be resolved, and this resolution involves the death of the neutrophils through the process of apoptosis. Although we know that neutrophils eventually die through apoptosis, the connection between neutrophil apoptosis and inflammation in bacterial infections has not been clear. In meningitis, bacteria gain access to the space surrounding the brain, multiply and cause inflammation. Despite antibiotic therapy, human pneumococcal meningitis often causes brain damage and death of the patient. We show here that the experimental inhibition of neutrophil apoptosis leads to much more severe clinical disease in a mouse model of bacterial (pneumococcal) meningitis. This was due to a prolonged inflammatory activity of the neutrophils. Experimental induction of neutrophil apoptosis in mice that had been infected and treated with antibiotics (mimicking human meningitis) improved the clinical condition and accelerated recovery. Experimental manipulation of neutrophil apoptosis can therefore be beneficial in acute inflammation.
Collapse
Affiliation(s)
- Uwe Koedel
- Department of Neurology, Clinic of the University of Munich, Munich, Germany
| | - Tobias Frankenberg
- Institute for Medical Microbiology, Technische Universität München, Munich, Germany
| | - Susanne Kirschnek
- Institute for Medical Microbiology, Technische Universität München, Munich, Germany
| | - Bianca Obermaier
- Department of Neurology, Clinic of the University of Munich, Munich, Germany
| | - Hans Häcker
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Robert Paul
- Department of Neurology, Clinic of the University of Munich, Munich, Germany
| | - Georg Häcker
- Institute for Medical Microbiology, Technische Universität München, Munich, Germany
- * E-mail:
| |
Collapse
|