1
|
Tian T, Xue Z, Sun X, Ding L, Zhao R, Wang Z, Wu J, Li X, Li Y, Sun L. Qingfei Yin alleviates Streptococcus pneumoniae pneumonia by promoting complete autophagy to suppress necroptosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 136:156280. [PMID: 39637472 DOI: 10.1016/j.phymed.2024.156280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/21/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Bacterial pneumonia is most prevalent among all pneumonia types, with Streptococcus pneumoniae being the main pathogen. Qingfei Yin (QFY) is a traditional Chinese medicine formula used in the clinical treatment of bacterial pneumonia. Previous studies have confirmed the multi-target and -effect characteristics of QFY in treating S. pneumoniae pneumonia. PURPOSE The purpose of this study was to explore the mechanism underlying QFY in treating pneumonia produced by S. pneumoniae. METHODS First, an in vivo model of S. pneumoniae-induced pneumonia was established in mice and evaluated the efficacy of QFY by hematoxylin-eosin (HE) staining and measuring cytokine levels in bronchoalveolar lavage fluid. Next, single-cell transcriptomics was used to identify the targeted cell subtypes, signaling pathways, and biological processes affected by QFY. Finally, the findings were validated using a pneumolysin (PLY) -induced mouse lung epithelial cells (TC-1) model in vitro using western blot analysis, immunofluorescence (IF), acridine orange (AO) staining, and propidium iodide (PI) staining. RESULTS QFY was shown to alleviate lung inflammation and reduce the TNF-α and IL-6 levels in bronchoalveolar lavage fluid in vivo. A total of 113,353 cells were classified using single-cell transcriptomics and 12 major cell types were identified. By single-cell transcriptomics, QFY was confirmed to primarily target lung epithelial cells. Differentially expressed genes were shown to be enriched in autophagy and necroptosis signaling pathways, and the key differentially expressed gene, Sequestosome 1 (p62/SQSTM1), was identified. PLY was shown to induce RIPK1-dependent necroptosis and incomplete autophagy in TC-1 cells. QFY was shown to promote complete autophagy by downregulating the expression of p62, thereby reducing phosphorylation of RIPK1 and MLKL, and alleviating necroptosis in S. pneumoniae-induced lung epithelial cell death. CONCLUSION This study demonstrated that QFY can effectively alleviate S. pneumoniae pneumonia. The mechanism of action may be that QFY promotes complete autophagy by downregulating p62 expression, thereby alleviating necroptosis of S. pneumoniae-induced lung epithelial cells and reducing lung injury. It provides a scientific basis for clinical prevention and treatment of S. pneumoniae pneumonia.
Collapse
Affiliation(s)
- Tong Tian
- Traditional Chinese Medicine College, Changchun University of Chinese Medicine, Changchun 130117, PR China
| | - Zhilong Xue
- Traditional Chinese Medicine College, Changchun University of Chinese Medicine, Changchun 130117, PR China
| | - Xiaozhou Sun
- Pediatric Respiratory Medicine Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, PR China
| | - Lizhong Ding
- Pediatric Respiratory Medicine Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, PR China
| | - Renshuang Zhao
- Medical College, Yanbian University, Yanji 133002, PR China
| | - Zhongtian Wang
- Traditional Chinese Medicine College, Changchun University of Chinese Medicine, Changchun 130117, PR China
| | - Jiaqi Wu
- Traditional Chinese Medicine College, Changchun University of Chinese Medicine, Changchun 130117, PR China
| | - Xiao Li
- Medical College, Yanbian University, Yanji 133002, PR China; Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun 130117, PR China.
| | - Yiquan Li
- Medical College, Yanbian University, Yanji 133002, PR China; Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun 130117, PR China.
| | - Liping Sun
- Pediatric Respiratory Medicine Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, PR China.
| |
Collapse
|
2
|
Anderson R, Feldman C. The Global Burden of Community-Acquired Pneumonia in Adults, Encompassing Invasive Pneumococcal Disease and the Prevalence of Its Associated Cardiovascular Events, with a Focus on Pneumolysin and Macrolide Antibiotics in Pathogenesis and Therapy. Int J Mol Sci 2023; 24:11038. [PMID: 37446214 DOI: 10.3390/ijms241311038] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/26/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Despite innovative advances in anti-infective therapies and vaccine development technologies, community-acquired pneumonia (CAP) remains the most persistent cause of infection-related mortality globally. Confronting the ongoing threat posed by Streptococcus pneumoniae (the pneumococcus), the most common bacterial cause of CAP, particularly to the non-immune elderly, remains challenging due to the propensity of the elderly to develop invasive pneumococcal disease (IPD), together with the predilection of the pathogen for the heart. The resultant development of often fatal cardiovascular events (CVEs), particularly during the first seven days of acute infection, is now recognized as a relatively common complication of IPD. The current review represents an update on the prevalence and types of CVEs associated with acute bacterial CAP, particularly IPD. In addition, it is focused on recent insights into the involvement of the pneumococcal pore-forming toxin, pneumolysin (Ply), in subverting host immune defenses, particularly the protective functions of the alveolar macrophage during early-stage disease. This, in turn, enables extra-pulmonary dissemination of the pathogen, leading to cardiac invasion, cardiotoxicity and myocardial dysfunction. The review concludes with an overview of the current status of macrolide antibiotics in the treatment of bacterial CAP in general, as well as severe pneumococcal CAP, including a consideration of the mechanisms by which these agents inhibit the production of Ply by macrolide-resistant strains of the pathogen.
Collapse
Affiliation(s)
- Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
| | - Charles Feldman
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand Medical School, 7 York Road, Johannesburg 2193, South Africa
| |
Collapse
|
3
|
Reyes LF, Garcia E, Ibáñez-Prada ED, Serrano-Mayorga CC, Fuentes YV, Rodríguez A, Moreno G, Bastidas A, Gómez J, Gonzalez A, Frei CR, Celi LA, Martin-Loeches I, Waterer G. Impact of macrolide treatment on long-term mortality in patients admitted to the ICU due to CAP: a targeted maximum likelihood estimation and survival analysis. Crit Care 2023; 27:212. [PMID: 37259125 DOI: 10.1186/s13054-023-04466-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/27/2023] [Indexed: 06/02/2023] Open
Abstract
INTRODUCTION Patients with community-acquired pneumonia (CAP) admitted to the intensive care unit (ICU) have high mortality rates during the acute infection and up to ten years thereafter. Recommendations from international CAP guidelines include macrolide-based treatment. However, there is no data on the long-term outcomes of this recommendation. Therefore, we aimed to determine the impact of macrolide-based therapy on long-term mortality in this population. METHODS Registered patients in the MIMIC-IV database 16 years or older and admitted to the ICU due to CAP were included. Multivariate analysis, targeted maximum likelihood estimation (TMLE) to simulate a randomised controlled trial, and survival analyses were conducted to test the effect of macrolide-based treatment on mortality six-month (6 m) and twelve-month (12 m) after hospital admission. A sensitivity analysis was performed excluding patients with Pseudomonas aeruginosa or MRSA pneumonia to control for Healthcare-Associated Pneumonia (HCAP). RESULTS 3775 patients were included, and 1154 were treated with a macrolide-based treatment. The non-macrolide-based group had worse long-term clinical outcomes, represented by 6 m [31.5 (363/1154) vs 39.5 (1035/2621), p < 0.001] and 12 m mortality [39.0 (450/1154) vs 45.7 (1198/2621), p < 0.001]. The main risk factors associated with long-term mortality were Charlson comorbidity index, SAPS II, septic shock, and respiratory failure. Macrolide-based treatment reduced the risk of dying at 6 m [HR (95% CI) 0.69 (0.60, 0.78), p < 0.001] and 12 m [0.72 (0.64, 0.81), p < 0.001]. After TMLE, the protective effect continued with an additive effect estimate of - 0.069. CONCLUSION Macrolide-based treatment reduced the hazard risk of long-term mortality by almost one-third. This effect remains after simulating an RCT with TMLE and the sensitivity analysis for the HCAP classification.
Collapse
Affiliation(s)
- Luis Felipe Reyes
- Universidad de La Sabana, Campus Puente del Común, KM 7.5 Autopista Norte de Bogotá, Chía, Colombia.
- Clínica Universidad de La Sabana, Chía, Colombia.
- University of Oxford, Oxford, UK.
| | - Esteban Garcia
- Universidad de La Sabana, Campus Puente del Común, KM 7.5 Autopista Norte de Bogotá, Chía, Colombia
| | | | | | - Yuli V Fuentes
- Universidad de La Sabana, Campus Puente del Común, KM 7.5 Autopista Norte de Bogotá, Chía, Colombia
- Clínica Universidad de La Sabana, Chía, Colombia
| | - Alejandro Rodríguez
- Hospital Universitari Joan XXIII, Critical Care Medicine, Rovira and Virgili University and CIBERES (Biomedical Research Network of Respiratory Disease), Tarragona, Spain
| | - Gerard Moreno
- Hospital Universitari Joan XXIII, Critical Care Medicine, Rovira and Virgili University and CIBERES (Biomedical Research Network of Respiratory Disease), Tarragona, Spain
| | - Alirio Bastidas
- Universidad de La Sabana, Campus Puente del Común, KM 7.5 Autopista Norte de Bogotá, Chía, Colombia
| | - Josep Gómez
- Hospital Universitari Joan XXIII, Critical Care Medicine, Rovira and Virgili University and CIBERES (Biomedical Research Network of Respiratory Disease), Tarragona, Spain
| | - Angélica Gonzalez
- Universidad de La Sabana, Campus Puente del Común, KM 7.5 Autopista Norte de Bogotá, Chía, Colombia
| | - Christopher R Frei
- College of Pharmacy, The University of Texas at Austin, San Antonio, TX, USA
- School of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Leo Anthony Celi
- Massachusetts Institute of Technology, Cambridge, USA
- Beth Israel Deaconess Medical Center, Boston, USA
- Harvard T.H. Chan School of Public Health, Boston, USA
| | - Ignacio Martin-Loeches
- Department of Intensive Care Medicine, Multidisciplinary Intensive Care Research Organisation (MICRO), St. James's Hospital, Dublin, Ireland
| | - Grant Waterer
- Royal Perth Bentley Hospital Group, University of Western Australia, Perth, Australia
| |
Collapse
|
4
|
Madzime M, Theron AJ, Anderson R, Tintinger GR, Steel HC, Meyer PWA, Nel JG, Feldman C, Rossouw TM. Dolutegravir potentiates platelet activation by a calcium-dependent, ionophore-like mechanism. J Immunotoxicol 2022; 19:1-8. [PMID: 36394569 DOI: 10.1080/1547691x.2022.2142705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Dolutegravir is a highly potent HIV integrase strand transfer inhibitor that is recommended for first-line anti-retroviral treatment in all major treatment guidelines. A recent study has shown that people taking this class of anti-retroviral treatment have a substantially higher risk of early-onset cardiovascular disease, a condition shown previously to be associated with increased platelet reactivity. To date, few studies have explored the effects of dolutegravir on platelet activation. Accordingly, the current study was undertaken with the primary objective of investigating the effects of dolutegravir on the reactivity of human platelets in vitro. Platelet-rich plasma, isolated platelets, or buffy coat cell suspensions prepared from the blood of healthy adults were treated with dolutegravir (2.5-10 µg/ml), followed by activation with adenosine 5'-diphosphate (ADP), thrombin, or a thromboxane A2 receptor agonist U46619. Expression of platelet CD62P (P-selectin), formation of heterotypic neutrophil:platelet aggregates, and calcium (Ca2+) fluxes were measured using flow cytometry and fluorescence spectrometry, respectively. Dolutegravir caused dose-related potentiation of ADP-, thrombin- and U46619-activated expression of CD62P by platelets, as well as a significant increases in formation of neutrophil:platelet aggregates. These effects were paralleled by a spontaneous, receptor-independent elevation in cytosolic Ca2+ that appears to underpin the mechanism by which the antiretroviral agent augments the responsiveness of these cells to ADP, thrombin and U46619. The most likely mechanism of dolutegravir-mediated increases in platelet cytosolic Ca2+ relates to a combination of lipophilicity and divalent/trivalent metal-binding and/or chelating properties of the anti-retroviral agent. These properties are likely to confer ionophore-type activities on dolutegravir that would promote movement of Ca2+ across the plasma membrane, delivering the cation to the cytosol where it would augment Ca2+-dependent intracellular signaling mechanisms. These effects of dolutegravir may lead to hyper-activation of platelets which, if operative in vivo, may contribute to an increased risk for cardiometabolic co-morbidities.
Collapse
Affiliation(s)
- Morris Madzime
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Annette J Theron
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Gregory R Tintinger
- Department of Internal Medicine, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Pretoria, South Africa
| | - Helen C Steel
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Pieter W A Meyer
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.,Department of Immunology, Tshwane Academic Division of the National Health Laboratory Service of South Africa, Pretoria, South Africa
| | - Jan G Nel
- Department of Haematology, Faculty of Health Sciences, University of Pretoria, and Tshwane Academic Division of the National Health Laboratory Service of South Africa, Pretoria, South Africa
| | - Charles Feldman
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Theresa M Rossouw
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
5
|
Sheng Q, Hou X, Wang N, Liu M, Zhu H, Deng X, Liang X, Chi G. Corilagin: A Novel Antivirulence Strategy to Alleviate Streptococcus pneumoniae Infection by Diminishing Pneumolysin Oligomers. Molecules 2022; 27:5063. [PMID: 36014299 PMCID: PMC9416474 DOI: 10.3390/molecules27165063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/30/2022] [Accepted: 08/05/2022] [Indexed: 11/17/2022] Open
Abstract
Pneumolysin (PLY) is a significant virulence factor of Streptococcus pneumoniae (S. pneumoniae), able to break through the defense system of a host and mediate the occurrence of a series of infections. Therefore, PLY as the most ideal target to prevent S. pneumoniae infection has received more and more attention and research. Corilagin is a tannic acid that exhibits excellent inhibition of PLY oligomers without bacteriostatic activity to S. pneumoniae. Herein, hemolytic activity assays, cell viability tests and western blot experiments are executed to evaluate the antivirulence efficacy of corilagin against PLY in vitro. Colony observation, hematoxylin and eosin (H&E) staining and cytokines of bronchoalveolar lavage fluid (BALF) are applied to assess the therapeutic effect of corilagin in mice infected by S. pneumoniae. The results indicate the related genes of corilagin act mainly via enrichment in pathways associated with pneumonia disease. Furthermore, molecular docking and molecular dynamics simulations show that corilagin might bind with domains 3 and 4 of PLY and interfere with its hemolytic activity, which is further confirmed by the site-directed mutagenesis of PLY. Additionally, corilagin limits PLY oligomer production without impacting PLY expression in S. pneumoniae cultures. Moreover, corilagin effectively relieves PLY-mediated cell injury without any cytotoxicity, even then reducing the colony count in the lung and the levels of pro-inflammatory factors in BALF and remarkably improving lung lesions. All the results demonstrate that corilagin may be a novel strategy to cope with S. pneumoniae infection by inhibiting PLY oligomerization.
Collapse
Affiliation(s)
- Qiushuang Sheng
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xiaoning Hou
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Nan Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Minda Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Haoyu Zhu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xuming Deng
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xiaoying Liang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
- Department of Internal Medicine, University of South Florida, Tampa, FL 33620, USA
| | - Gefu Chi
- The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010010, China
| |
Collapse
|
6
|
Zhang C, Deng Y, Wang X, Shi L, Zhan B, Hou N, Liu S, Bao M, Chi G, Fang T. Alnustone inhibits Streptococcus pneumoniae virulence by targeting pneumolysin and sortase A. Fitoterapia 2022; 162:105261. [DOI: 10.1016/j.fitote.2022.105261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/31/2022] [Accepted: 08/03/2022] [Indexed: 11/04/2022]
|
7
|
Jaiswal V, Ang SP, Lnu K, Ishak A, Pokhrel NB, Chia JE, Hajra A, Biswas M, Matetic A, Dhatt R, Mamas MA. Effect of Pneumococcal Vaccine on Mortality and Cardiovascular Outcomes: A Systematic Review and Meta-Analysis. J Clin Med 2022; 11:3799. [PMID: 35807082 PMCID: PMC9267914 DOI: 10.3390/jcm11133799] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/27/2022] [Accepted: 06/27/2022] [Indexed: 11/24/2022] Open
Abstract
Various studies have suggested the possible cardiovascular (CV) protective effects of the pneumococcal vaccine (PV). Therefore, we conducted a meta-analysis to assess the association between recipients of PV with mortality and CV outcomes among patients with and without established cardiovascular disease. We performed a systematic literature search in PubMed, Embase, and Scopus for studies evaluating the effect of PV on mortality and CV outcomes. A total of 15 studies with 347,444 patients were included in the meta-analysis: 111,784 patients received PV (32%) and 235,660 patients were in the unvaccinated group (68%). Recipients of PV were associated with decreased all-cause mortality (HR, 0.76 (95% CI: 0.66 to 0.87), p < 0.001). PV was associated with a decrease in the incidence of myocardial infarction (MI) (HR, 0.73 (95% CI: 0.56−0.96), p = 0.02), without significant reduction in CV mortality (HR, 0.87 (95% CI: 0.72−1.07), p = 0.18) and stroke (HR, 1.01 (95% CI: 0.93−1.10), p = 0.82). Our study found PV was associated with decreased risk of all-cause mortality and MI. Future RCTs will be necessary to confirm benefits associated with receipt of PV.
Collapse
Affiliation(s)
- Vikash Jaiswal
- Department of Medicine, Larkin Community Hospital, South Miami, FL 33143, USA;
| | - Song Peng Ang
- Department of Internal Medicine, Rutgers Health/Community Medical Center, Toms River, NJ 08755, USA;
| | - Kriti Lnu
- Department of Internal Medicine, UPMC Harrisburg, Harrisburg, PA 17105, USA;
| | - Angela Ishak
- School of Medicine, European University Cyprus, Nicosia 2404, Cyprus;
| | | | - Jia Ee Chia
- School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia;
| | - Adrija Hajra
- Department of Internal Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, The Bronx, NY 10461, USA;
| | - Monodeep Biswas
- Division of Cardiology, Wellspan Cardiology, Lancaster, PA 17602, USA;
| | - Andrija Matetic
- Department of Cardiology, University Hospital of Split, 21000 Split, Croatia;
- Keele Cardiovascular Research Group, Centre for Prognosis Research, Keele University, Keele ST5 5BG, UK
| | - Ravinder Dhatt
- Department of Internal Medicine, UPMC Harrisburg, Harrisburg, PA 17105, USA;
| | - Mamas A. Mamas
- Keele Cardiovascular Research Group, Centre for Prognosis Research, Keele University, Keele ST5 5BG, UK
| |
Collapse
|
8
|
Pereira JM, Xu S, Leong JM, Sousa S. The Yin and Yang of Pneumolysin During Pneumococcal Infection. Front Immunol 2022; 13:878244. [PMID: 35529870 PMCID: PMC9074694 DOI: 10.3389/fimmu.2022.878244] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/23/2022] [Indexed: 12/15/2022] Open
Abstract
Pneumolysin (PLY) is a pore-forming toxin produced by the human pathobiont Streptococcus pneumoniae, the major cause of pneumonia worldwide. PLY, a key pneumococcal virulence factor, can form transmembrane pores in host cells, disrupting plasma membrane integrity and deregulating cellular homeostasis. At lytic concentrations, PLY causes cell death. At sub-lytic concentrations, PLY triggers host cell survival pathways that cooperate to reseal the damaged plasma membrane and restore cell homeostasis. While PLY is generally considered a pivotal factor promoting S. pneumoniae colonization and survival, it is also a powerful trigger of the innate and adaptive host immune response against bacterial infection. The dichotomy of PLY as both a key bacterial virulence factor and a trigger for host immune modulation allows the toxin to display both "Yin" and "Yang" properties during infection, promoting disease by membrane perforation and activating inflammatory pathways, while also mitigating damage by triggering host cell repair and initiating anti-inflammatory responses. Due to its cytolytic activity and diverse immunomodulatory properties, PLY is integral to every stage of S. pneumoniae pathogenesis and may tip the balance towards either the pathogen or the host depending on the context of infection.
Collapse
Affiliation(s)
- Joana M. Pereira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Molecular and Cellular (MC) Biology PhD Program, ICBAS - Instituto de Ciência Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Shuying Xu
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, United States
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA, United States
| | - John M. Leong
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, United States
| | - Sandra Sousa
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| |
Collapse
|
9
|
Jahn K, Kohler TP, Swiatek LS, Wiebe S, Hammerschmidt S. Platelets, Bacterial Adhesins and the Pneumococcus. Cells 2022; 11:cells11071121. [PMID: 35406684 PMCID: PMC8997422 DOI: 10.3390/cells11071121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/22/2022] [Accepted: 03/22/2022] [Indexed: 01/25/2023] Open
Abstract
Systemic infections with pathogenic or facultative pathogenic bacteria are associated with activation and aggregation of platelets leading to thrombocytopenia and activation of the clotting system. Bacterial proteins leading to platelet activation and aggregation have been identified, and while platelet receptors are recognized, induced signal transduction cascades are still often unknown. In addition to proteinaceous adhesins, pathogenic bacteria such as Staphylococcus aureus and Streptococcus pneumoniae also produce toxins such as pneumolysin and alpha-hemolysin. They bind to cellular receptors or form pores, which can result in disturbance of physiological functions of platelets. Here, we discuss the bacteria-platelet interplay in the context of adhesin–receptor interactions and platelet-activating bacterial proteins, with a main emphasis on S. aureus and S. pneumoniae. More importantly, we summarize recent findings of how S. aureus toxins and the pore-forming toxin pneumolysin of S. pneumoniae interfere with platelet function. Finally, the relevance of platelet dysfunction due to killing by toxins and potential treatment interventions protecting platelets against cell death are summarized.
Collapse
|
10
|
Neutrophil-Derived Extracellular Vesicles Activate Platelets after Pneumolysin Exposure. Cells 2021; 10:cells10123581. [PMID: 34944089 PMCID: PMC8700313 DOI: 10.3390/cells10123581] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Pneumolysin (PLY) is a pore-forming toxin of Streptococcus pneumoniae that contributes substantially to the inflammatory processes underlying pneumococcal pneumonia and lung injury. Host responses against S. pneumoniae are regulated in part by neutrophils and platelets, both individually and in cooperative interaction. Previous studies have shown that PLY can target both neutrophils and platelets, however, the mechanisms by which PLY directly affects these cells and alters their interactions are not completely understood. In this study, we characterize the effects of PLY on neutrophils and platelets and explore the mechanisms by which PLY may induce neutrophil–platelet interactions. In vitro studies demonstrated that PLY causes the formation of neutrophil extracellular traps (NETs) and the release of extracellular vesicles (EVs) from both human and murine neutrophils. In vivo, neutrophil EV (nEV) levels were increased in mice infected with S. pneumoniae. In platelets, treatment with PLY induced the cell surface expression of P-selectin (CD62P) and binding to annexin V and caused a significant release of platelet EVs (pl-EVs). Moreover, PLY-induced nEVs but not NETs promoted platelet activation. The pretreatment of nEVs with proteinase K inhibited platelet activation, indicating that the surface proteins of nEVs play a role in this process. Our findings demonstrate that PLY activates neutrophils and platelets to release EVs and support an important role for neutrophil EVs in modulating platelet functions in pneumococcal infections.
Collapse
|
11
|
Pneumolysin induces platelet destruction, not platelet activation, which can be prevented by immunoglobulin preparations in vitro. Blood Adv 2021; 4:6315-6326. [PMID: 33351126 DOI: 10.1182/bloodadvances.2020002372] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 11/05/2020] [Indexed: 01/08/2023] Open
Abstract
Community-acquired pneumonia by primary or superinfections with Streptococcus pneumoniae can lead to acute respiratory distress requiring mechanical ventilation. The pore-forming toxin pneumolysin alters the alveolar-capillary barrier and causes extravasation of protein-rich fluid into the interstitial pulmonary tissue, which impairs gas exchange. Platelets usually prevent endothelial leakage in inflamed pulmonary tissue by sealing inflammation-induced endothelial gaps. We not only confirm that S pneumoniae induces CD62P expression in platelets, but we also show that, in the presence of pneumolysin, CD62P expression is not associated with platelet activation. Pneumolysin induces pores in the platelet membrane, which allow anti-CD62P antibodies to stain the intracellular CD62P without platelet activation. Pneumolysin treatment also results in calcium efflux, increase in light transmission by platelet lysis (not aggregation), loss of platelet thrombus formation in the flow chamber, and loss of pore-sealing capacity of platelets in the Boyden chamber. Specific anti-pneumolysin monoclonal and polyclonal antibodies inhibit these effects of pneumolysin on platelets as do polyvalent human immunoglobulins. In a post hoc analysis of the prospective randomized phase 2 CIGMA trial, we show that administration of a polyvalent immunoglobulin preparation was associated with a nominally higher platelet count and nominally improved survival in patients with severe S pneumoniae-related community-acquired pneumonia. Although, due to the low number of patients, no definitive conclusion can be made, our findings provide a rationale for investigation of pharmacologic immunoglobulin preparations to target pneumolysin by polyvalent immunoglobulin preparations in severe community-acquired pneumococcal pneumonia, to counteract the risk of these patients becoming ventilation dependent. This trial was registered at www.clinicaltrials.gov as #NCT01420744.
Collapse
|
12
|
Anderson R, Theron AJ, Steel HC, Nel JG, Tintinger GR. ADP-Mediated Upregulation of Expression of CD62P on Human Platelets Is Critically Dependent on Co-Activation of P2Y1 and P2Y12 Receptors. Pharmaceuticals (Basel) 2020; 13:ph13120420. [PMID: 33255391 PMCID: PMC7760858 DOI: 10.3390/ph13120420] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 12/17/2022] Open
Abstract
This study probed the differential utilization of P2Y1 and P2Y12 receptors in mobilizing CD62P (P-selectin) from intracellular granules following activation of human platelets with adenosine 5′-diphosphate (ADP, 100 µmol·L−1) Platelet-rich plasma (PRP) was prepared from the blood of adult humans. CD62P was measured by flow cytometry following activation of PRP with ADP in the absence and presence of the selective antagonists of P2Y1 and P2Y12 receptors, MRS2500 and PSB0739 (both 0.155–10 µmol·L−1), respectively. Effects of the test agents on ADP-activated, CD62P-dependent formation of neutrophil:platelet (NP) aggregates were also measured by flow cytometry, while phosphatidylinositol 3-kinase (PI3K) activity was measured according to Akt1 phosphorylation in platelet lysates. Treatment with MRS2500 or PSB0739 at 10 µmol·L−1 almost completely attenuated (94.6% and 86% inhibition, respectively) ADP-activated expression of CD62P and also inhibited NP aggregate formation. To probe the mechanisms involved in P2Y1/P2Y12 receptor-mediated expression of CD62P, PRP was pre-treated with U73122 (phospholipase C (PLC) inhibitor), 2-aminoethoxy-diphenyl borate (2-APB, inositol triphosphate receptor antagonist), calmidazolium chloride (calmodulin inhibitor), or wortmannin (PI3K inhibitor). U73122, 2-APB, and wortmannin caused almost complete inhibition of ADP-activated expression of CD62P, while calmidazolium chloride caused statistically significant, partial inhibition. PSB0739, but not MRS2500, caused potent inhibition of PI3K-mediated phosphorylation of Akt1. Optimal mobilization of CD62P by ADP-stimulated platelets is critically dependent on the co-activation of platelet P2Y1 and P2Y12 receptors. P2Y12 receptor activation is the key event in activation of PI3K, while activation of the P2Y1 receptor appears to create a high cytosolic Ca2+ environment conducive to optimum PI3K activity.
Collapse
Affiliation(s)
- Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (A.J.T.); (H.C.S.)
- Correspondence: ; Tel.: +27-12-318-2425; Fax: +27-12-323-0732
| | - Annette J. Theron
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (A.J.T.); (H.C.S.)
| | - Helen C. Steel
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (A.J.T.); (H.C.S.)
| | - Jan G. Nel
- Department of Haematology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
- Tshwane Academic Division of the National Laboratory Health Service of South Africa, Pretoria 0001, South Africa
| | - Gregory R. Tintinger
- Department of Internal Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
| |
Collapse
|
13
|
Panagiotou S, Chaguza C, Yahya R, Audshasai T, Baltazar M, Ressel L, Khandaker S, Alsahag M, Mitchell TJ, Prudhomme M, Kadioglu A, Yang M. Hypervirulent pneumococcal serotype 1 harbours two pneumolysin variants with differential haemolytic activity. Sci Rep 2020; 10:17313. [PMID: 33057054 PMCID: PMC7560715 DOI: 10.1038/s41598-020-73454-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/14/2020] [Indexed: 12/13/2022] Open
Abstract
Streptococcus pneumoniae is a devastating global pathogen. Prevalent in sub-Saharan Africa, pneumococcal serotype 1 is atypical in that it is rarely found as a nasopharyngeal coloniser, yet is described as one of the most common causes of invasive pneumococcal disease. Clonal sequence type (ST)-306 and ST615 are representative of the two major serotype 1 lineages A and C, respectively. Here we investigated the virulence properties and haemolytic activities of these 2 clonal types using in vivo mouse models and in vitro assays. A lethal dose of ST615 administered intranasally to mice led to the rapid onset of disease symptoms and resulted in 90% mortality. In contrast, mice exposed to the same infection dose of ST306 or a pneumolysin (Ply)-deficient ST615 failed to develop any disease symptoms. Interestingly, the 2 strains did not differ in their ability to bind the immune complement or to undergo neutrophil-mediated phagocytosis. Upon comparative genomic analysis, we found higher within-ST sequence diversity in ST615 compared with ST306 and determined that ZmpA, ZmpD proteins, and IgA protease, were uniquely found in ST615. Using cell fractionation and cell contact-dependent assay, we made the unexpected finding that ST615 harbours the expression of two haemolytic variants of Ply: a cell-wall restricted fully haemolytic Ply, and a cytosolic pool of Ply void of any detectable haemolytic activity. This is the first time such a phenomenon has been described. We discuss the biological significance of our observation in relation to the aptitude of the pneumococcus for sustaining its human reservoir.
Collapse
Affiliation(s)
- Stavros Panagiotou
- Department of Clinical Infection Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, The Ronald Ross Building, 8 West Derby St, Liverpool, L69 7BE, UK
| | - Chrispin Chaguza
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
- Darwin College, University of Cambridge, Silver Street, Cambridge, CB3 9EU, UK
| | - Reham Yahya
- College of sciences and health professions, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Teerawit Audshasai
- Department of Clinical Infection Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, The Ronald Ross Building, 8 West Derby St, Liverpool, L69 7BE, UK
| | - Murielle Baltazar
- Department of Clinical Infection Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, The Ronald Ross Building, 8 West Derby St, Liverpool, L69 7BE, UK
| | - Lorenzo Ressel
- Department of Veterinary Pathology and Public Health, Institute of Veterinary Science, University of Liverpool, Leahurst Campus, Neston, CH64 7TE, UK
| | - Shadia Khandaker
- Department of Clinical Infection Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, The Ronald Ross Building, 8 West Derby St, Liverpool, L69 7BE, UK
| | - Mansoor Alsahag
- Department of Clinical Infection Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, The Ronald Ross Building, 8 West Derby St, Liverpool, L69 7BE, UK
- Faculty of Applied Medical Sciences, Albaha University, Albaha, Kingdom of Saudi Arabia
| | - Tim J Mitchell
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Marc Prudhomme
- Université Paul Sabatier, Centre National de la Recherche Scientifique, 118 Route de Narbonne, 31062, Toulouse Cedex 9, France
| | - Aras Kadioglu
- Department of Clinical Infection Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, The Ronald Ross Building, 8 West Derby St, Liverpool, L69 7BE, UK.
| | - Marie Yang
- Department of Clinical Infection Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, The Ronald Ross Building, 8 West Derby St, Liverpool, L69 7BE, UK.
| |
Collapse
|
14
|
Feldman C, Anderson R. Platelets and Their Role in the Pathogenesis of Cardiovascular Events in Patients With Community-Acquired Pneumonia. Front Immunol 2020; 11:577303. [PMID: 33042161 PMCID: PMC7527494 DOI: 10.3389/fimmu.2020.577303] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022] Open
Abstract
Community-acquired pneumonia (CAP) remains an important cause of morbidity and mortality throughout the world with much recent and ongoing research focused on the occurrence of cardiovascular events (CVEs) during the infection, which are associated with adverse short-term and long-term survival. Much of the research directed at unraveling the pathogenesis of these events has been undertaken in the settings of experimental and clinical CAP caused by the dangerous, bacterial respiratory pathogen, Streptococcus pneumoniae (pneumococcus), which remains the most common bacterial cause of CAP. Studies of this type have revealed that although platelets play an important role in host defense against infection, there is also increasing recognition that hyperactivation of these cells contributes to a pro-inflammatory, prothrombotic systemic milieu that contributes to the etiology of CVEs. In the case of the pneumococcus, platelet-driven myocardial damage and dysfunction is exacerbated by the direct cardiotoxic actions of pneumolysin, a major pore-forming toxin of this pathogen, which also acts as potent activator of platelets. This review is focused on the role of platelets in host defense against infection, including pneumococcal infection in particular, and reviews the current literature describing the potential mechanisms by which platelet activation contributes to cardiovascular complications in CAP. This is preceded by an evaluation of the burden of pneumococcal infection in CAP, the clinical features and putative pathogenic mechanisms of the CVE, and concludes with an evaluation of the potential utility of the anti-platelet activity of macrolides and various adjunctive therapies.
Collapse
Affiliation(s)
- Charles Feldman
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, Institute of Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
15
|
Nishimoto AT, Rosch JW, Tuomanen EI. Pneumolysin: Pathogenesis and Therapeutic Target. Front Microbiol 2020; 11:1543. [PMID: 32714314 PMCID: PMC7343714 DOI: 10.3389/fmicb.2020.01543] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 06/15/2020] [Indexed: 01/08/2023] Open
Abstract
Streptococcus pneumoniae is an opportunistic pathogen responsible for widespread illness and is a major global health issue for children, the elderly, and the immunocompromised population. Pneumolysin (PLY) is a cholesterol-dependent cytolysin (CDC) and key pneumococcal virulence factor involved in all phases of pneumococcal disease, including transmission, colonization, and infection. In this review we cover the biology and cytolytic function of PLY, its contribution to S. pneumoniae pathogenesis, and its known interactions and effects on the host with regard to tissue damage and immune response. Additionally, we review statins as a therapeutic option for CDC toxicity and PLY toxoid as a vaccine candidate in protein-based vaccines.
Collapse
Affiliation(s)
- Andrew T Nishimoto
- Department of Infectious Disease, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Jason W Rosch
- Department of Infectious Disease, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Elaine I Tuomanen
- Department of Infectious Disease, St. Jude Children's Research Hospital, Memphis, TN, United States
| |
Collapse
|
16
|
Wolff M, Handtke S, Palankar R, Wesche J, Kohler TP, Kohler C, Gruel Y, Hammerschmidt S, Greinacher A. Activated platelets kill Staphylococcus aureus, but not Streptococcus pneumoniae-The role of FcγRIIa and platelet factor 4/heparinantibodies. J Thromb Haemost 2020; 18:1459-1468. [PMID: 32237268 DOI: 10.1111/jth.14814] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 03/16/2020] [Accepted: 03/23/2020] [Indexed: 02/01/2023]
Abstract
BACKGROUND Heparin induced thrombocytopenia (HIT) is likely a misdirected bacterial host defense mechanism. Platelet factor 4 (PF4) binds to polyanions on bacterial surfaces exposing neo-epitopes to which HIT antibodies bind. Platelets are activated by the resulting immune complexes via FcγRIIA, release bactericidal substances, and kill Gram-negative Escherichia coli. OBJECTIVES To assess the role of PF4, anti-PF4/H antibodies and FcγRIIa in killing of Gram-positive bacteria by platelets. METHODS Binding of PF4 to protein-A deficient Staphylococcus aureus (SA113Δspa) and non-encapsulated Streptococcus pneumoniae (D39Δcps) and its conformational change were assessed by flow cytometry using monoclonal (KKO,5B9) and patient derived anti-PF4/H antibodies. Killing of bacteria was quantified by counting colony forming units (cfu) after incubation with platelets or platelet releasate. Using flow cytometry, platelet activation (CD62P-expression, PAC-1 binding) and phosphatidylserine (PS)-exposure were analyzed. RESULTS Monoclonal and patient-derived anti-PF4/H antibodies bound in the presence of PF4 to both S. aureus and S. pneumoniae (1.6-fold increased fluorescence signal for human anti-PF4/H antibodies to 24.0-fold increase for KKO). Staphylococcus aureus (5.5 × 104 cfu/mL) was efficiently killed by platelets (2.7 × 104 cfu/mL) or their releasate (2.9 × 104 cfu/mL). Killing was not further enhanced by PF4 or anti-PF4/H antibodies. Blocking FcγRIIa had no impact on killing of S. aureus by platelets. In contrast, S. pneumoniae was not killed by platelets or releasate. Instead, after incubation with pneumococci platelets were unresponsive to TRAP-6 stimulation and exposed high levels of PS. CONCLUSIONS Anti-PF4/H antibodies seem to have only a minor role for direct killing of Gram-positive bacteria by platelets. Staphylococcus aureus is killed by platelets or platelet releasate. In contrast, S. pneumoniae affects platelet viability.
Collapse
Affiliation(s)
- Martina Wolff
- Institut für Immunologie und Transfusion Medizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Stefan Handtke
- Institut für Immunologie und Transfusion Medizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Raghavendra Palankar
- Institut für Immunologie und Transfusion Medizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Jan Wesche
- Institut für Immunologie und Transfusion Medizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Thomas P Kohler
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Christian Kohler
- Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Yves Gruel
- Département d'Hématologie-Hémostase, Hôpital Universitaire de Tours, Tours, France
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Andreas Greinacher
- Institut für Immunologie und Transfusion Medizin, Universitätsmedizin Greifswald, Greifswald, Germany
| |
Collapse
|
17
|
Feldman C. Cardiac complications in community-acquired pneumonia and COVID-19. Afr J Thorac Crit Care Med 2020; 26:10.7196/AJTCCM.2020.v26i2.077. [PMID: 34235421 PMCID: PMC7221539 DOI: 10.7196/ajtccm.2020.v26i2.077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2020] [Indexed: 12/11/2022] Open
Abstract
Community-acquired pneumonia (CAP) remains a global health problem with significant morbidity and mortality. Much recent published literature about the infection has indicated that a substantial number of patients with CAP, particularly those ill enough to be admitted to hospital, will suffer a cardiovascular event. While these may include events such as deep venous thrombosis and stroke, most of the events involve the heart and include the occurrence of an arrhythmia (most commonly atrial fibrillation), new onset or worsening of heart failure and acute myocardial infarction. While such cardiac events may occur, for example, in all-cause CAP and CAP due to influenza virus infection, and more recently described with the SARS-CoV-2 pandemic, a significant amount of research work has been investigating the pathogenic mechanisms of these cardiac events in patients with CAP due to Streptococcus pneumoniae (pneumococcus) and, more recently, COVID-19 infections. Such research has identified a number of mechanisms by which these microorganisms may cause cardiovascular events. Importantly, these cardiac events appear not only to be associated with in-hospital mortality, but they also appear to contribute to longer-term mortality of patients with CAP, even after their discharge from hospital. This review will focus initially on studies of cardiovascular events in all-cause CAP and pneumococcal CAP, excluding COVID-19 infection, and then address similar issues in the latter infection.
Collapse
Affiliation(s)
- C Feldman
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
18
|
|
19
|
Severiche-Bueno D, Parra-Tanoux D, Reyes LF, Waterer GW. Hot topics and current controversies in community-acquired pneumonia. Breathe (Sheff) 2019; 15:216-225. [PMID: 31508159 PMCID: PMC6717612 DOI: 10.1183/20734735.0205-2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Community-acquired pneumonia (CAP) is one of the most common infectious diseases, as well as a major cause of death both in developed and developing countries, and it remains a challenge for physicians around the world. Several guidelines have been published to guide clinicians in how to diagnose and take care of patients with CAP. However, there are still many areas of debate and uncertainty where research is needed to advance patient care and improve clinical outcomes. In this review we highlight current hot topics in CAP and present updated evidence around these areas of controversy. Community-acquired pneumonia is the most frequent cause of infectious death worldwide; however, there are several areas of controversy that should be addressed to improve patient care. This review presents the available data on these topics.http://bit.ly/2ShnH7A
Collapse
Affiliation(s)
- Diego Severiche-Bueno
- Infectious Diseases and Critical Care Depts, Universidad de La Sabana, Chía, Colombia
| | - Daniela Parra-Tanoux
- Infectious Diseases and Critical Care Depts, Universidad de La Sabana, Chía, Colombia
| | - Luis F Reyes
- Infectious Diseases and Critical Care Depts, Universidad de La Sabana, Chía, Colombia
| | - Grant W Waterer
- Royal Perth Bentley Hospital Group, University of Western Australia, Perth, Australia
| |
Collapse
|
20
|
Binsker U, Kohler TP, Hammerschmidt S. Contribution of Human Thrombospondin-1 to the Pathogenesis of Gram-Positive Bacteria. J Innate Immun 2019; 11:303-315. [PMID: 30814475 PMCID: PMC6738282 DOI: 10.1159/000496033] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 12/03/2018] [Indexed: 12/12/2022] Open
Abstract
A successful colonization of different compartments of the human host requires multifactorial contacts between bacterial surface proteins and host factors. Extracellular matrix proteins and matricellular proteins such as thrombospondin-1 play a pivotal role as adhesive substrates to ensure a strong interaction with pathobionts like the Gram-positive Streptococcus pneumoniae and Staphylococcus aureus. The human glycoprotein thrombospondin-1 is a component of the extracellular matrix and is highly abundant in the bloodstream during bacteremia. Human platelets secrete thrombospondin-1, which is then acquired by invading pathogens to facilitate colonization and immune evasion. Gram-positive bacteria express a broad spectrum of surface-exposed proteins, some of which also recognize thrombospondin-1. This review highlights the importance of thrombospondin-1 as an adhesion substrate to facilitate colonization, and we summarize the variety of thrombospondin-1-binding proteins of S. pneumoniae and S. aureus.
Collapse
Affiliation(s)
- Ulrike Binsker
- Center for Functional Genomics of Microbes, Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Greifswald University, Greifswald, Germany
- Department of Microbiology, NYU Langone Health, Alexandria Center for the Life Sciences, New York City, New York, USA
| | - Thomas P Kohler
- Center for Functional Genomics of Microbes, Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Greifswald University, Greifswald, Germany
| | - Sven Hammerschmidt
- Center for Functional Genomics of Microbes, Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, Greifswald University, Greifswald, Germany,
| |
Collapse
|
21
|
Anderson R, Theron AJ, Nel JG, Durandt C, Cholo MC, Feldman C, Tintinger GR. Clofazimine, but Not Isoniazid or Rifampicin, Augments Platelet Activation in vitro. Front Pharmacol 2018; 9:1335. [PMID: 30515097 PMCID: PMC6255828 DOI: 10.3389/fphar.2018.01335] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 10/30/2018] [Indexed: 12/22/2022] Open
Abstract
Although the inclusion of the cationic amphiphilic, anti-mycobacterial agent, clofazimine, in the chemotherapeutic regimens of patients with multidrug-resistant tuberculosis (TB) has contributed to improved outcomes, concerns remain about the cardiotoxic potential of this agent. Accordingly, the current study was undertaken with the primary objective of investigating the effects of clofazimine, on the reactivity of human platelets in vitro, a seemingly unexplored, mechanism of cardiotoxicity. Platelet-rich plasma (PRP) prepared from the blood of healthy, adult humans was treated with clofazimine (0.625–10 mg/L), or the primary anti-TB agents, isoniazid and rifampicin (at final concentrations of 5 and 10 mg/L), followed by addition of either adenosine 5′-diphosphate (ADP) or thrombin and measurement of platelet activation according to the magnitude of expression of CD62P (P-selectin), as well as the CD62P-mediated formation of heterotypic neutrophil:platelet (NP) aggregates, using flow cytometry. Clofazimine, but neither isoniazid nor rifampicin, caused dose-related potentiation of both ADP- and thrombin-activated expression of CD62P by platelets, achieving statistical significance at threshold concentrations of 0.625 and 2.5 mg/L, respectively, as well as significant formation of N:P aggregates. These stimulatory effects of clofazimine on platelet activation were partly attenuated by pre-treatment of PRP with the membrane-stabilizing agent, α-tocopherol, possibly consistent with a membrane-disruptive mechanism. In conclusion, clofazimine, at concentrations within the therapeutic range, augments platelet activation in vitro, probably by a mechanism linked to membrane destabilization. If operative in vivo, these pro-thrombotic activities of clofazimine may predispose for development of microvascular occlusion, exacerbating an already existing high risk for development of TB-associated cardiovascular disease.
Collapse
Affiliation(s)
- Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.,Institute for Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Annette J Theron
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Jan G Nel
- Department of Haematology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.,Tshwane Academic Division, National Health Laboratory Service, Pretoria, South Africa
| | - Chrisna Durandt
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.,Institute for Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Moloko C Cholo
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Charles Feldman
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Gregory R Tintinger
- Department of Internal Medicine, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
22
|
Zhang W, Wang H, Wang B, Zhang Y, Hu Y, Ma B, Wang J. Replacing the 238th aspartic acid with an arginine impaired the oligomerization activity and inflammation-inducing property of pyolysin. Virulence 2018; 9:1112-1125. [PMID: 30067143 PMCID: PMC6086297 DOI: 10.1080/21505594.2018.1491256] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Trueperella pyogenes (T. pyogenes) is an important opportunistic pathogen. Pyolysin (PLO) importantly contributes to the pathogenicity of T. pyogenes. However, the relationship between the structure and function and the virulence of PLO is not well documented. In the current study, recombinant PLO (rPLO) and three rPLO mutants were prepared. rPLO D238R, a mutant with the 238th aspartic acid replaced with an arginine, showed impairment in oligomerization activity on cholesterol-containing liposome and pore-forming activity on sheep red blood cell membrane. Further study employing the prepared mutants confirmed that the pore-forming activity of PLO is essential for inducing excessive inflammation responses in mice by upregulating the expression levels of IL-1β, TNF-α, and IL-6. By contrast, rPLO P499F, another mutant with impaired cell membrane binding capacity, elicited an inflammation response that was dependent on pathogen-associated molecular pattern (PAMP) activity, given that the mutant significantly upregulated the expression of IL-10 in macrophages and in mice, whereas rPLO did not. Results indicated that domain 1 of the PLO molecule plays an important role in maintaining pore-forming activity. Moreover, the PLO pore-forming activity and not PAMP activity is responsible for the inflammation-inducing effect of PLO. The results of this study provided new information for research field on the structure, function, and virulence of PLO. Abbreviations: T. pyogenes: Trueperella pyogenes; PLO: Pyolysin; rPLO: recombinant PLO; PAMP: pathogen-associated molecular pattern; CDCs: cholesterol-dependent cytolysins; PLY: pneumolysin; NLRP3: NLR family pyrin domain containing protein 3; PRRs: pattern recognition receptors; Asp: aspartic acid; TLR4: Toll-like receptor 4; Arg: arginine; Asn: asparagine; IPTG: Isopropyl-β-d-thiogalactoside; PBS: phosphate-buffered saline; sRBCs: sheep red blood cells; TEM: Transmission electron microscopy; RBCM: red blood cell membrane; SDS-PAGE: sodium dodecyl sulfate–polyacrylamide gel electrophoresis; NC membrane: nitrocellulose membrane; SDS-AGE: dodecyl sulfate agarose gel electrophoresis; MDBK cells: Madin–Darby bovine kidney cells; RPMI-1640 medium: Roswell Park Memorial Institute-1640 medium; FBS: fetal bovine serum; BMDMs: bone marrow-derived macrophages; TNF-α: tumor necrosis factor α; IL-1β: interleukin-1β; IFN-γ: interferon-γ; TGF-β: transforming growth factor-β; ELISA: enzyme-linked immunosorbent assay
Collapse
Affiliation(s)
- Wenlong Zhang
- a College of Veterinary Medicine , Northeast Agricultural University , Harbin , PR China.,b China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology , Northeastern Science Inspection Station , Harbin , PR China
| | - Haili Wang
- a College of Veterinary Medicine , Northeast Agricultural University , Harbin , PR China.,b China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology , Northeastern Science Inspection Station , Harbin , PR China
| | - Bing Wang
- a College of Veterinary Medicine , Northeast Agricultural University , Harbin , PR China.,b China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology , Northeastern Science Inspection Station , Harbin , PR China
| | - Yue Zhang
- a College of Veterinary Medicine , Northeast Agricultural University , Harbin , PR China.,b China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology , Northeastern Science Inspection Station , Harbin , PR China
| | - Yunhao Hu
- a College of Veterinary Medicine , Northeast Agricultural University , Harbin , PR China.,b China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology , Northeastern Science Inspection Station , Harbin , PR China
| | - Bo Ma
- a College of Veterinary Medicine , Northeast Agricultural University , Harbin , PR China.,b China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology , Northeastern Science Inspection Station , Harbin , PR China
| | - Junwei Wang
- a College of Veterinary Medicine , Northeast Agricultural University , Harbin , PR China.,b China Ministry of Agriculture Key Laboratory of Animal Pathogen Biology , Northeastern Science Inspection Station , Harbin , PR China
| |
Collapse
|
23
|
Bouillot S, Reboud E, Huber P. Functional Consequences of Calcium Influx Promoted by Bacterial Pore-Forming Toxins. Toxins (Basel) 2018; 10:toxins10100387. [PMID: 30257425 PMCID: PMC6215193 DOI: 10.3390/toxins10100387] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 09/14/2018] [Accepted: 09/20/2018] [Indexed: 02/06/2023] Open
Abstract
Bacterial pore-forming toxins induce a rapid and massive increase in cytosolic Ca2+ concentration due to the formation of pores in the plasma membrane and/or activation of Ca2+-channels. As Ca2+ is an essential messenger in cellular signaling, a sustained increase in Ca2+ concentration has dramatic consequences on cellular behavior, eventually leading to cell death. However, host cells have adapted mechanisms to protect against Ca2+ intoxication, such as Ca2+ efflux and membrane repair. The final outcome depends upon the nature and concentration of the toxin and on the cell type. This review highlights the repercussions of Ca2+ overload on the induction of cell death, repair mechanisms, cellular adhesive properties, and the inflammatory response.
Collapse
Affiliation(s)
- Stéphanie Bouillot
- Université Grenoble Alpes, CNRS ERL5261, CEA BIG-BCI, INSERM UMR1036, Grenoble 38054, France.
| | - Emeline Reboud
- Université Grenoble Alpes, CNRS ERL5261, CEA BIG-BCI, INSERM UMR1036, Grenoble 38054, France.
| | - Philippe Huber
- Université Grenoble Alpes, CNRS ERL5261, CEA BIG-BCI, INSERM UMR1036, Grenoble 38054, France.
| |
Collapse
|
24
|
Anderson R, Nel JG, Feldman C. Multifaceted Role of Pneumolysin in the Pathogenesis of Myocardial Injury in Community-Acquired Pneumonia. Int J Mol Sci 2018; 19:E1147. [PMID: 29641429 PMCID: PMC5979279 DOI: 10.3390/ijms19041147] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/14/2018] [Accepted: 03/21/2018] [Indexed: 12/14/2022] Open
Abstract
Pneumolysin (PLY), a member of the family of Gram-positive bacterial, cholesterol-dependent, β-barrel pore-forming cytolysins, is the major protein virulence factor of the dangerous respiratory pathogen, Streptococcus pneumoniae (pneumococcus). PLY plays a major role in the pathogenesis of community-acquired pneumonia (CAP), promoting colonization and invasion of the upper and lower respiratory tracts respectively, as well as extra-pulmonary dissemination of the pneumococcus. Notwithstanding its role in causing acute lung injury in severe CAP, PLY has also been implicated in the development of potentially fatal acute and delayed-onset cardiovascular events, which are now recognized as being fairly common complications of this condition. This review is focused firstly on updating mechanisms involved in the immunopathogenesis of PLY-mediated myocardial damage, specifically the direct cardiotoxic and immunosuppressive activities, as well as the indirect pro-inflammatory/pro-thrombotic activities of the toxin. Secondly, on PLY-targeted therapeutic strategies including, among others, macrolide antibiotics, natural product antagonists, cholesterol-containing liposomes, and fully humanized monoclonal antibodies, as well as on vaccine-based preventive strategies. These sections are preceded by overviews of CAP in general, the role of the pneumococcus as the causative pathogen, the occurrence and types of CAP-associated cardiac complication, and the structure and biological activities of PLY.
Collapse
Affiliation(s)
- Ronald Anderson
- Department of Immunology and Institute for Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa.
| | - Jan G Nel
- Department of Haematology, Faculty of Health Sciences, University of Pretoria and Tshwane Academic Division of the National Health Laboratory Service, Pretoria 0001, South Africa.
| | - Charles Feldman
- Division of Pulmonology, Department of Internal Medicine, Charlotte Maxeke Johannesburg Academic Hospital and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 0002, South Africa.
| |
Collapse
|
25
|
Restrepo MI, Reyes LF. Pneumonia as a cardiovascular disease. Respirology 2018; 23:250-259. [PMID: 29325222 DOI: 10.1111/resp.13233] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 08/05/2017] [Indexed: 01/08/2023]
Abstract
Community-acquired pneumonia (CAP) is an important cause of death around the globe. Up to 30% of patients admitted to hospital for CAP develop cardiovascular complications (i.e. new/worsening heart failure, new/worsening arrhythmias, myocardial infarctions and/or strokes), acutely and up to 10 years thereafter. Cardiac complications result from complex interactions between preexisting conditions, relative ischaemia, upregulation of the sympathetic system, systemic inflammation and direct pathogen-mediated damage to the cardiovascular system. The exact mechanisms underlying the direct host-pathogen interactions are of great interest to identify potential therapeutic and preventative targets for CAP. In this review, we summarize the epidemiological data, risk factors and the pathogen-driven cardiovascular damage affecting patients with CAP.
Collapse
Affiliation(s)
- Marcos I Restrepo
- Division of Pulmonary Diseases and Critical Care Medicine, South Texas Veterans Health Care System, San Antonio, TX, USA.,Department of Medicine, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Luis F Reyes
- Division of Pulmonary Diseases and Critical Care Medicine, South Texas Veterans Health Care System, San Antonio, TX, USA.,Department of Medicine, University of Texas Health at San Antonio, San Antonio, TX, USA
| |
Collapse
|
26
|
Anderson R, Feldman C. Review manuscript: Mechanisms of platelet activation by the pneumococcus and the role of platelets in community-acquired pneumonia. J Infect 2017; 75:473-485. [PMID: 28943342 DOI: 10.1016/j.jinf.2017.09.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 09/13/2017] [Accepted: 09/15/2017] [Indexed: 12/11/2022]
Abstract
There is increasing recognition of the involvement of platelets in orchestrating inflammatory responses, driving the activation of neutrophils, monocytes and vascular endothelium, which, if poorly controlled, may lead to microvascular dysfunction. Importantly, hyperreactivity of platelets has been implicated in the pathogenesis of myocardial injury and the associated particularly high prevalence of acute cardiovascular events in patients with severe community-acquired pneumonia (CAP), of which Streptococcus pneumoniae (pneumococcus) is the most commonly encountered aetiologic agent. In this context, it is noteworthy that a number of studies have documented various mechanisms by which the pneumococcus may directly promote platelet aggregation and activation. The major contributors to platelet activation include several different types of pneumococcal adhesin, the pore-forming toxin, pneumolysin, and possibly pathogen-derived hydrogen peroxide, which collectively represent a major focus of the current review. This is followed by an overview of the limited experimental studies together with a larger series of clinical studies mainly focused on all-cause CAP, which have provided evidence in support of associations between alterations in circulating platelet counts, most commonly thrombocytopenia, and a poor clinical outcome. The final section of the review covers, albeit briefly, systemic biomarkers of platelet activation which may have prognostic potential.
Collapse
Affiliation(s)
- Ronald Anderson
- Department of Immunology and Institute for Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.
| | - Charles Feldman
- Division of Pulmonology, Department of Internal Medicine, Charlotte Maxeke Johannesburg Academic Hospital and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
27
|
Song M, Teng Z, Li M, Niu X, Wang J, Deng X. Epigallocatechin gallate inhibits Streptococcus pneumoniae virulence by simultaneously targeting pneumolysin and sortase A. J Cell Mol Med 2017; 21:2586-2598. [PMID: 28402019 PMCID: PMC5618700 DOI: 10.1111/jcmm.13179] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 02/24/2017] [Indexed: 01/11/2023] Open
Abstract
Streptococcus pneumoniae (pneumococcus), the causative agent of several human diseases, possesses numerous virulence factors associated with pneumococcal infection and pathogenesis. Pneumolysin (PLY), an important virulence factor, is a member of the cholesterol-dependent cytolysin family and has cytolytic activity. Sortase A (SrtA), another crucial pneumococcal virulence determinate, contributes greatly to the anchoring of many virulence-associated surface proteins to the cell wall. In this study, epigallocatechin gallate (EGCG), a natural compound with little known antipneumococcal activity, was shown to directly inhibit PLY-mediated haemolysis and cytolysis by blocking the oligomerization of PLY and simultaneously reduce the peptidase activity of SrtA. The biofilm formation, production of neuraminidase A (NanA, the pneumococcal surface protein anchored by SrtA), and bacterial adhesion to human epithelial cells (Hep2) were inhibited effectively when S. pneumoniae D39 was cocultured with EGCG. The results from molecular dynamics simulations and mutational analysis confirmed the interaction of EGCG with PLY and SrtA, and EGCG binds to Glu277, Tyr358, and Arg359 in PLY and Thr169, Lys171, and Phe239 in SrtA. In vivo studies further demonstrated that EGCG protected mice against S. pneumoniae pneumonia. Our results imply that EGCG is an effective inhibitor of both PLY and SrtA and that an antivirulence strategy that directly targets PLY and SrtA using EGCG is a promising therapeutic option for S. pneumoniae pneumonia.
Collapse
Affiliation(s)
- Meng Song
- The First Hospital and Institute of Infection and Immunity, Jilin University, Changchun, China.,Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zihao Teng
- The First Hospital and Institute of Infection and Immunity, Jilin University, Changchun, China.,Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Meng Li
- The First Hospital and Institute of Infection and Immunity, Jilin University, Changchun, China.,Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiaodi Niu
- The First Hospital and Institute of Infection and Immunity, Jilin University, Changchun, China.,Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jianfeng Wang
- The First Hospital and Institute of Infection and Immunity, Jilin University, Changchun, China.,Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xuming Deng
- The First Hospital and Institute of Infection and Immunity, Jilin University, Changchun, China
| |
Collapse
|
28
|
Anderson R, Feldman C. Pneumolysin as a potential therapeutic target in severe pneumococcal disease. J Infect 2017; 74:527-544. [PMID: 28322888 DOI: 10.1016/j.jinf.2017.03.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/09/2017] [Accepted: 03/11/2017] [Indexed: 12/13/2022]
Abstract
Acute pulmonary and cardiac injury remain significant causes of morbidity and mortality in those afflicted with severe pneumococcal disease, with the risk for early mortality often persisting several years beyond clinical recovery. Although remaining to be firmly established in the clinical setting, a considerable body of evidence, mostly derived from murine models of experimental infection, has implicated the pneumococcal, cholesterol-binding, pore-forming toxin, pneumolysin (Ply), in the pathogenesis of lung and myocardial dysfunction. Topics covered in this review include the burden of pneumococcal disease, risk factors, virulence determinants of the pneumococcus, complications of severe disease, antibiotic and adjuvant therapies, as well as the structure of Ply and the role of the toxin in disease pathogenesis. Given the increasing recognition of the clinical potential of Ply-neutralisation strategies, the remaining sections of the review are focused on updates of the types, benefits and limitations of currently available therapies which may attenuate, directly and/or indirectly, the injurious actions of Ply. These include recently described experimental therapies such as various phytochemicals and lipids, and a second group of more conventional agents the members of which remain the subject of ongoing clinical evaluation. This latter group, which is covered more extensively, encompasses macrolides, statins, corticosteroids, and platelet-targeted therapies, particularly aspirin.
Collapse
Affiliation(s)
- Ronald Anderson
- Department of Immunology and Institute of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.
| | - Charles Feldman
- Division of Pulmonology, Department of Internal Medicine, Charlotte Maxeke Johannesburg Academic Hospital and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
29
|
Nel JG, Durandt C, Theron AJ, Tintinger GR, Pool R, Richards GA, Mitchell TJ, Feldman C, Anderson R. Pneumolysin mediates heterotypic aggregation of neutrophils and platelets in vitro. J Infect 2017; 74:599-608. [PMID: 28267572 DOI: 10.1016/j.jinf.2017.02.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/17/2017] [Accepted: 02/24/2017] [Indexed: 10/20/2022]
Abstract
OBJECTIVES Platelets orchestrate the inflammatory activities of neutrophils, possibly contributing to pulmonary and myocardial damage during severe pneumococcal infection. This study tested the hypothesis that the pneumococcal toxin, pneumolysin (Ply), activates production of platelet-activating factor (PAF) and thromboxane A2 (TxA2) by neutrophils, these bioactive lipids being potential mediators of neutrophil:platelet (NP) networking. METHODS The effects of recombinant Ply (10-80 ng mL-1) on the production of PAF and TxA2 by isolated neutrophils were measured using ELISA procedures, and NP aggregation by flow cytometry. RESULTS Exposure of neutrophils to Ply induced production of PAF and, to a lesser extent, TxA2, achieving statistical significance at ≥20 ng mL-1 of the toxin. In the case of NP interactions, Ply promoted heterotypic aggregation which was dependent on upregulation of P-selectin (CD62P) and activation of protease-activated receptor 1 (PAR1), attaining statistical significance at ≥10 ng mL-1 of the toxin, but did not involve either PAF or TxA2. CONCLUSION Ply induces synthesis of PAF and TxA2, by human neutrophils, neither of which appears to contribute to the formation of NP heterotypic aggregates in vitro, a process which is seemingly dependent on CD62P and PAR1. These pro-inflammatory activities of Ply may contribute to the pathogenesis of pulmonary and myocardial injury during severe pneumococcal infection.
Collapse
Affiliation(s)
- Jan G Nel
- Department of Haematology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa; Tshwane Academic Division of the National Health Laboratory Service, Pretoria, South Africa.
| | - Chrisna Durandt
- Institute for Cellular and Molecular Medicine, South African Medical Research Council Unit for Stem Cell Research, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Annette J Theron
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Gregory R Tintinger
- Department of Internal Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Roger Pool
- Department of Haematology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa; Tshwane Academic Division of the National Health Laboratory Service, Pretoria, South Africa
| | - Guy A Richards
- Department of Critical Care, Charlotte Maxeke Johannesburg Academic Hospital, Johannesburg, South Africa; Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Timothy J Mitchell
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Charles Feldman
- Division of Pulmonology, Department of Internal Medicine, Charlotte Maxeke Johannesburg Academic Hospital, Johannesburg, South Africa
| | - Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa; Institute for Cellular and Molecular Medicine, South African Medical Research Council Unit for Stem Cell Research, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
30
|
Prise en charge des infections à Streptococcus pneumoniae : ce qui a changé en 15 ans. MEDECINE INTENSIVE REANIMATION 2017. [DOI: 10.1007/s13546-017-1265-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
31
|
Abstract
Pneumococcal infections continue to cause significant morbidity and mortality in patients throughout the world. This microorganism remains the most common bacterial cause of community-acquired pneumonia and is associated with a considerable burden of disease and health-care costs in both developed and developing countries. Emerging antibiotic resistance has been a concern because of its potential negative impact on the outcome of patients who receive standard antibiotic therapy. However, there have been substantial changes in the epidemiology of this pathogen in recent years, not least of which has been due to the use of pneumococcal conjugate vaccines in children, with subsequent herd protection in unvaccinated adults and children. Furthermore, much recent research has led to a better understanding of the virulence factors of this pathogen and their role in the pathogenesis of severe pneumococcal disease, including the cardiac complications, as well as the potential role of adjunctive therapy in the management of severely ill cases. This review will describe recent advances in our understanding of the epidemiology, virulence factors, and management of pneumococcal community-acquired pneumonia.
Collapse
Affiliation(s)
- Charles Feldman
- Charlotte Maxeke Johannesburg Academic Hospital and Faculty of Health Sciences, University of the Witwatersrand Medical School, Johannesburg, South Africa
| | - Ronald Anderson
- Institute for Cellular and Molecular Medicine, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
32
|
Feldman C, Anderson R. Prevalence, pathogenesis, therapy, and prevention of cardiovascular events in patients with community-acquired pneumonia. Pneumonia (Nathan) 2016; 8:11. [PMID: 28702290 PMCID: PMC5471702 DOI: 10.1186/s41479-016-0011-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 07/04/2016] [Indexed: 12/11/2022] Open
Abstract
It is now well recognised that cardiac events occur relatively commonly in patients with acute community-acquired pneumonia. While these events are more frequent in patients with underlying risk factors—such as those with underlying chronic cardiovascular and respiratory comorbidities, the elderly, and in nursing home residents—they also occur in patients with no underlying risks other than severe pneumonia. Recent research elucidating the underlying pathogenic mechanisms related to these cardiac events has indicated a probable role for platelet activation, which is possibly exacerbated by pneumolysin in the case of pneumococcal infections. This, in turn, has resulted in the identification of possible therapeutic strategies targeting platelet activation, as well as the cardio-toxic activity of pneumolysin. These issues represent the primary focus of the current review.
Collapse
Affiliation(s)
- Charles Feldman
- Division of Pulmonology, Department of Internal Medicine, Charlotte Maxeke Johannesburg Academic Hospital, Johannesburg, South Africa.,Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Department of Internal Medicine, University of the Witwatersrand Medical School, 7 York Road, Parktown, 2193 Johannesburg, South Africa
| | - Ronald Anderson
- Institute for Cellular and Molecular Medicine, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|