1
|
Hollwedel FD, Maus R, Stolper J, Iwai S, Kasai H, Holtfreter S, Pich A, Neubert L, Welte T, Yamasaki S, Maus UA. Ectopic Expression of C-Type Lectin Mincle Renders Mice Susceptible to Staphylococcal Pneumonia. J Infect Dis 2024; 230:198-208. [PMID: 39052710 DOI: 10.1093/infdis/jiad608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/05/2023] [Accepted: 12/29/2023] [Indexed: 01/03/2024] Open
Abstract
Staphylococcus aureus is a prevalent pathogen in pneumonia and harbors glycolipids, which may serve as molecular patterns in Mincle (macrophage-inducible C-type lectin)-dependent pathogen recognition. We examined the role of Mincle in lung defense against S aureus in wild-type (WT), Mincle knockout (KO), and Mincle transgenic (tg) mice. Two glycolipids, glucosyl-diacylglycerol (Glc-DAG) and diglucosyl-diacylglycerol (Glc2-DAG), were purified, of which only Glc-DAG triggered Mincle reporter cell activation and professional phagocyte responses. Proteomic profiling revealed that Glc2-DAG blocked Glc-DAG-induced cytokine responses, thereby acting as inhibitor of Glc-DAG/Mincle signaling. WT mice responded to S aureus with a similar lung pathology as Mincle KO mice, most likely due to Glc2-DAG-dependent inhibition of Glc-DAG/Mincle signaling. In contrast, ectopic Mincle expression caused severe lung pathology in S aureus-infected mice, characterized by bacterial outgrowth and fatal pneumonia. Collectively, Glc2-DAG inhibits Glc-DAG/Mincle-dependent responses in WT mice, whereas sustained Mincle expression overrides Glc2-DAG-mediated inhibitory effects, conferring increased host susceptibility to S aureus.
Collapse
Affiliation(s)
- Femke D Hollwedel
- Division of Experimental Pneumology, Hannover Medical School, Hannover, Germany
| | - Regina Maus
- Division of Experimental Pneumology, Hannover Medical School, Hannover, Germany
| | - Jennifer Stolper
- Division of Experimental Pneumology, Hannover Medical School, Hannover, Germany
| | - Satoru Iwai
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Hayato Kasai
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Silva Holtfreter
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Andreas Pich
- Institute of Toxicology and Core Facility Proteomics, Hannover Medical School, Hannover, Germany
| | - Lavinia Neubert
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Tobias Welte
- Clinic for Pneumology, Hannover Medical School, Hannover, Germany
- German Center for Lung Research, partner site BREATH, Hannover, Germany
| | - Sho Yamasaki
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Ulrich A Maus
- Division of Experimental Pneumology, Hannover Medical School, Hannover, Germany
- German Center for Lung Research, partner site BREATH, Hannover, Germany
| |
Collapse
|
2
|
Pan T, Lee JW. A crucial role of neutrophil extracellular traps in pulmonary infectious diseases. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:34-41. [PMID: 39170960 PMCID: PMC11332830 DOI: 10.1016/j.pccm.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Indexed: 08/23/2024]
Abstract
Neutrophil extracellular traps (NETs), extrusions of intracellular DNA with attached granular material that exert an antibacterial effect through entangling, isolating, and immobilizing microorganisms, have been extensively studied in recent decades. The primary role of NETs is to entrap and facilitate the killing of bacteria, fungi, viruses, and parasites, preventing bacterial and fungal dissemination. NET formation has been described in many pulmonary diseases, including both infectious and non-infectious. NETs are considered a double-edged sword. As innate immune cells, neutrophils release NETs to kill pathogens and remove cellular debris. However, the deleterious effects of excessive NET release in lung disease are particularly important because NETs and by-products of NETosis can directly induce epithelial and endothelial cell death while simultaneously inducing inflammatory cytokine secretion and immune-mediated thrombosis. Thus, NET formation must be tightly regulated to preserve the anti-microbial capability of NETs while minimizing damage to the host. In this review, we summarized the recent updates on the mechanism of NETs formation and pathophysiology associated with excessive NETs, aiming to provide insights for research and treatment of pulmonary infectious diseases.
Collapse
Affiliation(s)
- Ting Pan
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jae Woo Lee
- Department of Anesthesiology, University of California Los Angeles, Los Angeles, CA 90230, USA
| |
Collapse
|
3
|
Xu S, Xiong Y, Fu B, Guo D, Sha Z, Lin X, Wu H. Bacteria and macrophages in the tumor microenvironment. Front Microbiol 2023; 14:1115556. [PMID: 36825088 PMCID: PMC9941202 DOI: 10.3389/fmicb.2023.1115556] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 01/12/2023] [Indexed: 02/10/2023] Open
Abstract
Cancer and microbial infections are significant worldwide health challenges. Numerous studies have demonstrated that bacteria may contribute to the emergence of cancer. In this review, we assemble bacterial species discovered in various cancers to describe their variety and specificity. The relationship between bacteria and macrophages in cancer is also highlighted, and we look for ample proof to establish a biological basis for bacterial-induced macrophage polarization. Finally, we quickly go over the potential roles of metabolites, cytokines, and microRNAs in the regulation of the tumor microenvironment by bacterially activated macrophages. The complexity of bacteria and macrophages in cancer will be revealed as we gain a better understanding of their pathogenic mechanisms, which will lead to new therapeutic approaches for both inflammatory illnesses and cancer.
Collapse
Affiliation(s)
| | | | - Beibei Fu
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Dong Guo
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Zhou Sha
- School of Life Sciences, Chongqing University, Chongqing, China
| | | | | |
Collapse
|
4
|
Palmer CS, Kimmey JM. Neutrophil Recruitment in Pneumococcal Pneumonia. Front Cell Infect Microbiol 2022; 12:894644. [PMID: 35646729 PMCID: PMC9136017 DOI: 10.3389/fcimb.2022.894644] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 04/18/2022] [Indexed: 01/19/2023] Open
Abstract
Streptococcus pneumoniae (Spn) is the primary agent of community-acquired pneumonia. Neutrophils are innate immune cells that are essential for bacterial clearance during pneumococcal pneumonia but can also do harm to host tissue. Neutrophil migration in pneumococcal pneumonia is therefore a major determinant of host disease outcomes. During Spn infection, detection of the bacterium leads to an increase in proinflammatory signals and subsequent expression of integrins and ligands on both the neutrophil as well as endothelial and epithelial cells. These integrins and ligands mediate the tethering and migration of the neutrophil from the bloodstream to the site of infection. A gradient of host-derived and bacterial-derived chemoattractants contribute to targeted movement of neutrophils. During pneumococcal pneumonia, neutrophils are rapidly recruited to the pulmonary space, but studies show that some of the canonical neutrophil migratory machinery is dispensable. Investigation of neutrophil migration is necessary for us to understand the dynamics of pneumococcal infection. Here, we summarize what is known about the pathways that lead to migration of the neutrophil from the capillaries to the lung during pneumococcal infection.
Collapse
Affiliation(s)
| | - Jacqueline M. Kimmey
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, Santa Cruz, CA, United States
| |
Collapse
|
5
|
Li H, Bradbury JA, Edin ML, Graves JP, Gruzdev A, Cheng J, Hoopes SL, DeGraff LM, Fessler MB, Garantziotis S, Schurman SH, Zeldin DC. sEH promotes macrophage phagocytosis and lung clearance of Streptococcus pneumoniae. J Clin Invest 2021; 131:129679. [PMID: 34591792 DOI: 10.1172/jci129679] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 09/28/2021] [Indexed: 12/12/2022] Open
Abstract
Epoxyeicosatrienoic acids (EETs) have potent antiinflammatory properties. Hydrolysis of EETs by soluble epoxide hydrolase/ epoxide hydrolase 2 (sEH/EPHX2) to less active diols attenuates their antiinflammatory effects. Macrophage activation is critical to many inflammatory responses; however, the role of EETs and sEH in regulating macrophage function remains unknown. Lung bacterial clearance of Streptococcus pneumoniae was impaired in Ephx2-deficient (Ephx2-/-) mice and in mice treated with an sEH inhibitor. The EET receptor antagonist EEZE restored lung clearance of S. pneumoniae in Ephx2-/- mice. Ephx2-/- mice had normal lung Il1b, Il6, and Tnfa expression levels and macrophage recruitment to the lungs during S. pneumoniae infection; however, Ephx2 disruption attenuated proinflammatory cytokine induction, Tlr2 and Pgylrp1 receptor upregulation, and Ras-related C3 botulinum toxin substrates 1 and 2 (Rac1/2) and cell division control protein 42 homolog (Cdc42) activation in PGN-stimulated macrophages. Consistent with these observations, Ephx2-/- macrophages displayed reduced phagocytosis of S. pneumoniae in vivo and in vitro. Heterologous overexpression of TLR2 and peptidoglycan recognition protein 1 (PGLYRP1) in Ephx2-/- macrophages restored macrophage activation and phagocytosis. Human macrophage function was similarly regulated by EETs. Together, these results demonstrate that EETs reduced macrophage activation and phagocytosis of S. pneumoniae through the downregulation of TLR2 and PGLYRP1 expression. Defining the role of EETs and sEH in macrophage function may lead to the development of new therapeutic approaches for bacterial diseases.
Collapse
|
6
|
Herta T, Bhattacharyya A, Rosolowski M, Conrad C, Gurtner C, Gruber AD, Ahnert P, Gutbier B, Frey D, Suttorp N, Hippenstiel S, Zahlten J. Krueppel-Like Factor 4 Expression in Phagocytes Regulates Early Inflammatory Response and Disease Severity in Pneumococcal Pneumonia. Front Immunol 2021; 12:726135. [PMID: 34589087 PMCID: PMC8473698 DOI: 10.3389/fimmu.2021.726135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/25/2021] [Indexed: 11/13/2022] Open
Abstract
The transcription factor Krueppel-like factor (KLF) 4 fosters the pro-inflammatory immune response in macrophages and polymorphonuclear neutrophils (PMNs) when stimulated with Streptococcus pneumoniae, the main causative pathogen of community-acquired pneumonia (CAP). Here, we investigated the impact of KLF4 expression in myeloid cells such as macrophages and PMNs on inflammatory response and disease severity in a pneumococcal pneumonia mouse model and in patients admitted to hospital with CAP. We found that mice with a myeloid-specific knockout of KLF4 mount an insufficient early immune response with reduced levels of pro-inflammatory cytokines and increased levels of the anti-inflammatory cytokine interleukin (IL) 10 in bronchoalveolar lavage fluid and plasma and an impaired bacterial clearance from the lungs 24 hours after infection with S. pneumoniae. This results in higher rates of bacteremia, increased lung tissue damage, more severe symptoms of infection and reduced survival. Higher KLF4 gene expression levels in the peripheral blood of patients with CAP at hospital admission correlate with a favourable clinical presentation (lower sequential organ failure assessment (SOFA) score), lower serum levels of IL-10 at admission, shorter hospital stay and lower mortality or requirement of intensive care unit treatment within 28 days after admission. Thus, KLF4 in myeloid cells such as macrophages and PMNs is an important regulator of the early pro-inflammatory immune response and, therefore, a potentially interesting target for therapeutic interventions in pneumococcal pneumonia.
Collapse
Affiliation(s)
- Toni Herta
- Department of Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Aritra Bhattacharyya
- Department of Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Maciej Rosolowski
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, Leipzig, Germany
| | - Claudia Conrad
- Department of Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Corinne Gurtner
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Achim D. Gruber
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Peter Ahnert
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, Leipzig, Germany
| | - Birgitt Gutbier
- Department of Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Doris Frey
- Department of Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Norbert Suttorp
- Department of Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Hippenstiel
- Department of Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Janine Zahlten
- Department of Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
7
|
Domon H, Terao Y. The Role of Neutrophils and Neutrophil Elastase in Pneumococcal Pneumonia. Front Cell Infect Microbiol 2021; 11:615959. [PMID: 33796475 PMCID: PMC8008068 DOI: 10.3389/fcimb.2021.615959] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 03/01/2021] [Indexed: 12/11/2022] Open
Abstract
Streptococcus pneumoniae, also known as pneumococcus, is a Gram-positive diplococcus and a major human pathogen. This bacterium is a leading cause of bacterial pneumonia, otitis media, meningitis, and septicemia, and is a major cause of morbidity and mortality worldwide. To date, studies on S. pneumoniae have mainly focused on the role of its virulence factors including toxins, cell surface proteins, and capsules. However, accumulating evidence indicates that in addition to these studies, knowledge of host factors and host-pathogen interactions is essential for understanding the pathogenesis of pneumococcal diseases. Recent studies have demonstrated that neutrophil accumulation, which is generally considered to play a critical role in host defense during bacterial infections, can significantly contribute to lung injury and immune subversion, leading to pneumococcal invasion of the bloodstream. Here, we review bacterial and host factors, focusing on the role of neutrophils and their elastase, which contribute to the progression of pneumococcal pneumonia.
Collapse
Affiliation(s)
- Hisanori Domon
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Research Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yutaka Terao
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Research Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
8
|
Nickol ME, Lyle SM, Dennehy B, Kindrachuk J. Dysregulated Host Responses Underlie 2009 Pandemic Influenza-Methicillin Resistant Staphylococcus aureus Coinfection Pathogenesis at the Alveolar-Capillary Barrier. Cells 2020; 9:E2472. [PMID: 33202895 PMCID: PMC7696554 DOI: 10.3390/cells9112472] [Citation(s) in RCA: 3] [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: 10/17/2020] [Revised: 11/11/2020] [Accepted: 11/11/2020] [Indexed: 02/04/2023] Open
Abstract
Influenza viruses are a continual public health concern resulting in 3-5 million severe infections annually despite intense vaccination campaigns and messaging. Secondary bacterial infections, including Staphylococcus aureus, result in increased morbidity and mortality during seasonal epidemics and pandemics. While coinfections can result in deleterious pathologic consequences, including alveolar-capillary barrier disruption, the underlying mechanisms are poorly understood. We have characterized host- and pathogen-centric mechanisms contributing to influenza-bacterial coinfections in a primary cell coculture model of the alveolar-capillary barrier. Using 2009 pandemic influenza (pH1N1) and methicillin-resistant S. aureus (MRSA), we demonstrate that coinfection resulted in dysregulated barrier function. Preinfection with pH1N1 resulted in modulation of adhesion- and invasion-associated MRSA virulence factors during lag phase bacterial replication. Host response modulation in coinfected alveolar epithelial cells were primarily related to TLR- and inflammatory response-mediated cell signaling events. While less extensive in cocultured endothelial cells, coinfection resulted in changes to cellular stress response- and TLR-related signaling events. Analysis of cytokine expression suggested that cytokine secretion might play an important role in coinfection pathogenesis. Taken together, we demonstrate that coinfection pathogenesis is related to complex host- and pathogen-mediated events impacting both epithelial and endothelial cell regulation at the alveolar-capillary barrier.
Collapse
Affiliation(s)
- Michaela E. Nickol
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (M.E.N.); (S.M.L.); (B.D.)
| | - Sarah M. Lyle
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (M.E.N.); (S.M.L.); (B.D.)
| | - Brendan Dennehy
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (M.E.N.); (S.M.L.); (B.D.)
| | - Jason Kindrachuk
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (M.E.N.); (S.M.L.); (B.D.)
- Vaccine and Infectious Disease Organization-International Vaccine Centre, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| |
Collapse
|
9
|
Disruption of the cpsE and endA Genes Attenuates Streptococcus pneumoniae Virulence: Towards the Development of a Live Attenuated Vaccine Candidate. Vaccines (Basel) 2020; 8:vaccines8020187. [PMID: 32326482 PMCID: PMC7349068 DOI: 10.3390/vaccines8020187] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/27/2020] [Accepted: 04/07/2020] [Indexed: 12/15/2022] Open
Abstract
The majority of deaths due to Streptococcus pneumoniae infections are in developing countries. Although polysaccharide-based pneumococcal vaccines are available, newer types of vaccines are needed to increase vaccine affordability, particularly in developing countries, and to provide broader protection across all pneumococcal serotypes. To attenuate pneumococcal virulence with the aim of engineering candidate live attenuated vaccines (LAVs), we constructed knockouts in S. pneumoniae D39 of one of the capsular biosynthetic genes, cpsE that encodes glycosyltransferase, and the endonuclease gene, endA, that had been implicated in the uptake of DNA from the environment as well as bacterial escape from neutrophil-mediated killing. The cpsE gene knockout significantly lowered peak bacterial density, BALB/c mice nasopharyngeal (NP) colonisation but increased biofilm formation when compared to the wild-type D39 strain as well as the endA gene knockout mutant. All constructed mutant strains were able to induce significantly high serum and mucosal antibody response in BALB/c mice. However, the cpsE-endA double mutant strain, designated SPEC, was able to protect mice from high dose mucosal challenge of the D39 wild-type. Furthermore, SPEC showed 23-fold attenuation of virulence compared to the wild-type. Thus, the cpsE-endA double-mutant strain could be a promising candidate for further development of a LAV for S. pneumoniae.
Collapse
|
10
|
García López E, Martín-Galiano AJ. The Versatility of Opportunistic Infections Caused by Gemella Isolates Is Supported by the Carriage of Virulence Factors From Multiple Origins. Front Microbiol 2020; 11:524. [PMID: 32296407 PMCID: PMC7136413 DOI: 10.3389/fmicb.2020.00524] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 03/11/2020] [Indexed: 12/29/2022] Open
Abstract
The molecular basis of the pathogenesis of the opportunistic invasive infections caused by isolates of the Gemella genus remains largely unknown. Moreover, inconsistencies in the current species assignation were detected after genome-level comparison of 16 public Gemella isolates. A literature search detected that, between the two most pathogenic species, Gemella morbillorum causes about twice the number of cases compared to Gemella haemolysans. These two species shared their mean diseases - sepsis and endocarditis - but differed in causing other syndromes. A number of well-known virulence factors were harbored by all species, such as a manganese transport/adhesin sharing 83% identity from oral endocarditis-causing streptococci. Likewise, all Gemellae carried the genes required for incorporating phosphorylcholine into their cell walls and encoded some choline-binding proteins. In contrast, other proteins were species-specific, which may justify the known epidemiological differences. G. haemolysans, but not G. morbillorum, harbor a gene cluster potentially encoding a polysaccharidic capsule. Species-specific surface determinants also included Rib and MucBP repeats, hemoglobin-binding NEAT domains, peptidases of C5a complement factor and domains that recognize extracellular matrix molecules exposed in damaged heart valves, such as collagen and fibronectin. Surface virulence determinants were associated with several taxonomically dispersed opportunistic genera of the oral microbiota, such as Granulicatella, Parvimonas, and Streptococcus, suggesting the existence of a horizontally transferrable gene reservoir in the oral environment, likely facilitated by close proximity in biofilms and ultimately linked to endocarditis. The identification of the Gemella virulence pool should be implemented in whole genome-based protocols to rationally predict the pathogenic potential in ongoing clinical infections caused by these poorly known bacterial pathogens.
Collapse
Affiliation(s)
- Ernesto García López
- Departamento de Biotecnología Microbiana y de Plantas, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Antonio J. Martín-Galiano
- Intrahospital Infections Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III (ISCIII), Majadahonda, Spain
| |
Collapse
|
11
|
Kim JY, Seo SM, Lee HK, Kim HW, Choi YK. Comparison of the virulence of Streptococcus pneumoniae in ICR mouse stocks of three different origins. Lab Anim Res 2019; 35:5. [PMID: 31463224 PMCID: PMC6707427 DOI: 10.1186/s42826-019-0002-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 04/11/2019] [Indexed: 01/13/2023] Open
Abstract
Streptococcus pneumoniae causes many people to suffer from pneumonia, septicemia, and other diseases worldwide. To identify the difference in susceptibility of and treatment efficacy against S. pneumoniae in three ICR mouse stocks (Korl:ICR, A:ICR, and B:ICR) with different origins, mice were infected with 2 × 106, 2 × 107, and 2 × 108 CFU of S. pneumoniae D39 intratracheally. The survival of mice was observed until three weeks after the infection. The three stocks of mice showed no significant survival rate difference at 2 × 106 and 2 × 107 CFU. However, the lung and spleen weight in the A:ICR stock was significantly different from that in the other two stocks, whereas the liver weight in B:ICR stock was significantly lower than that in the other two stocks. Interestingly, no significant CFU difference in the organs was observed between the ICR stocks. The level of interferon gamma inducible protein 10 in Korl:ICR was significantly lower than that in the other two stocks. The level of granulocyte colony stimulating factor in B:ICR was significantly lower than in the other two stocks. However, tumor-necrosis factor-alpha and interleukin-6 levels showed no significant difference between the ICR stocks. In the vancomycin efficacy test after the S. pneumoniae infection, both the single-dose and double-dose vancomycin-treated groups showed a significantly better survival rate than the control group. There was no significant survival difference between the three stocks. These data showed that Korl:ICR, A:ICR, and B:ICR have no susceptibility difference to the S. pneumoniae D39 serotype 2.
Collapse
Affiliation(s)
- Jun-Young Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029 Republic of Korea
| | - Sun-Min Seo
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029 Republic of Korea
| | - Han-Kyul Lee
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029 Republic of Korea
| | - Han-Woong Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029 Republic of Korea
| | - Yang-Kyu Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029 Republic of Korea
| |
Collapse
|
12
|
Topical application of nebulized human IgG, IgA and IgAM in the lungs of rats and non-human primates. Respir Res 2019; 20:99. [PMID: 31118031 PMCID: PMC6532128 DOI: 10.1186/s12931-019-1057-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/24/2019] [Indexed: 01/31/2023] Open
Abstract
Background Recurrent and persistent infections are known to affect airways of patients with Primary Immunodeficiency despite appropriate replacement immunoglobulin serum levels. Interestingly, patients with Chronic Obstructive Pulmonary Disease or with non-CF bronchiectasis also show similar susceptibility to such infections. This may be due to the limited availability of immunoglobulins from the systemic circulation in the conductive airways, resulting in local immunodeficiency. Topical application of nebulized plasma-derived immunoglobulins may represent a means to address this deficiency. In this study, we assessed the feasibility of nebulizing plasma-derived immunoglobulins and delivering them into the airways of rats and non-human primates. Methods Distinct human plasma-derived immunoglobulin isotype preparations were nebulized with an investigational eFlow® nebulizer and analyzed in vitro or deposited into animals. Biochemical and immunohistological analysis of nebulized immunoglobulins were then performed. Lastly, efficacy of topically applied human plasma-derived immunoglobulins was assessed in an acute Streptococcus pneumoniae respiratory infection in mice. Results Characteristics of the resulting aerosols were comparable between preparations, even when using solutions with elevated viscosity. Neither the structural integrity nor the biological function of nebulized immunoglobulins were compromised by the nebulization process. In animal studies, immunoglobulins levels were assessed in plasma, broncho-alveolar lavages (BAL) and on lung sections of rats and non-human primates in samples collected up to 72 h following application. Nebulized immunoglobulins were detectable over 48 h in the BAL samples and up to 72 h on lung sections. Immunoglobulins recovered from BAL fluid up to 24 h after inhalation remained structurally and functionally intact. Importantly, topical application of human plasma-derived immunoglobulin G into the airways of mice offered significant protection against acute pneumococcal pneumonia. Conclusion Taken together our data demonstrate the feasibility of topically applying plasma-derived immunoglobulins into the lungs using a nebulized liquid formulation. Moreover, topically administered human plasma-derived immunoglobulins prevented acute respiratory infection. Electronic supplementary material The online version of this article (10.1186/s12931-019-1057-3) contains supplementary material, which is available to authorized users.
Collapse
|
13
|
Palomino-Segura M, Gonzalez SF. Two-Photon Intravital Imaging of Leukocytes in the Trachea During Pneumococcal Infection. Methods Mol Biol 2019; 1968:183-194. [PMID: 30929215 DOI: 10.1007/978-1-4939-9199-0_15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Two-photon intravital imaging (2P-IVM) of the murine trachea is a powerful technique for real-time imaging of immune cell recruitment and trafficking during airborne pathogen infections. Neutrophils are an important component of the innate immune response that are able to rapidly infiltrate the airway mucosa in response to Streptococcus pneumoniae infection. Here we describe a protocol to visualize in vivo neutrophil extravasation and cell dynamics in the tracheal tissue of a S. pneumoniae-infected mouse using 2P-IVM. To perform this protocol, we infected and imaged the trachea of a lysozyme M green fluorescent protein (LysM-GFP) mouse, in which neutrophils express GFP. Additionally, we used a custom-designed platform, which allowed the intubation and fixation of the trachea after surgical exposition, and we injected intravenously a fluorescently labeled dextran solution to visualize the blood vessels.
Collapse
Affiliation(s)
- Miguel Palomino-Segura
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Graduate School of Cellular and Molecular Sciences, Faculty of Medicine, University of Bern, Bern, Switzerland
| | - Santiago F Gonzalez
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland.
| |
Collapse
|
14
|
Desialylation of Platelets by Pneumococcal Neuraminidase A Induces ADP-Dependent Platelet Hyperreactivity. Infect Immun 2018; 86:IAI.00213-18. [PMID: 30037798 DOI: 10.1128/iai.00213-18] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 07/13/2018] [Indexed: 11/20/2022] Open
Abstract
Platelets are increasingly recognized to play a role in the complications of Streptococcus pneumoniae infections. S. pneumoniae expresses neuraminidases, which may alter glycans on the platelet surface. In the present study, we investigated the capability of pneumococcal neuraminidase A (NanA) to remove sialic acid (desialylation) from the platelet surface, the consequences for the platelet activation status and reactivity, and the ability of neuraminidase inhibitors to prevent these effects. Our results show that soluble NanA induces platelet desialylation. Whereas desialylation itself did not induce platelet activation (P-selectin expression and platelet fibrinogen binding), platelets became hyperreactive to ex vivo stimulation by ADP and cross-linked collagen-related peptide (CRP-XL). Platelet aggregation with leukocytes also increased. These processes were dependent on the ADP pathway, as inhibitors of the pathway (apyrase and ticagrelor) abrogated platelet hyperreactivity. Inhibition of NanA-induced platelet desialylation by neuraminidase inhibitors (e.g., oseltamivir acid) also prevented the platelet effects of NanA. Collectively, our findings show that soluble NanA can desialylate platelets, leading to platelet hyperreactivity, which can be prevented by neuraminidase inhibitors.
Collapse
|
15
|
Jwa MY, Ko EB, Kim HY, Kim SK, Jeong S, Seo HS, Yun CH, Han SH. Gamma-irradiation-killed Streptococcus pneumoniae potently induces the expression of IL-6 and IL-8 in human bronchial epithelial cells. Microb Pathog 2018; 124:38-46. [PMID: 30114464 DOI: 10.1016/j.micpath.2018.08.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/10/2018] [Accepted: 08/13/2018] [Indexed: 12/18/2022]
Abstract
Streptococcus pneumoniae is a major respiratory pathogen that can cause pneumonia, meningitis, and otitis media. Although capsular polysaccharide-based vaccines are commercially available, there is a need for broad-spectrum, serotype-independent, and cost-effective vaccines. Recently, an intranasal vaccine formulated with gamma-irradiated nonencapsulated S. pneumoniae whole cells has been developed and its immunogenicity is under investigation. Since innate immunity influences the subsequent adaptive immunity, in the present study, we investigated the immunostimulatory activity of gamma-irradiated S. pneumoniae (r-SP) in the human bronchial epithelial cell-line, BEAS-2B, by comparing with heat-inactivated S. pneumoniae (h-SP) and formalin-inactivated S. pneumoniae (f-SP). r-SP potently induced interleukin (IL)-6 and IL-8 at both mRNA and protein levels in a dose- and time-dependent manner, whereas h-SP and f-SP poorly induced them. Of note, the mRNA levels of IL-6 and IL-8 were approximately two-fold higher when cells were stimulated with 3 × 107 CFU/ml of r-SP for 3 h, while the protein levels of IL-6 and IL-8 were approximately five-fold higher after stimulation with 3 × 107 CFU/ml of r-SP for 24 h. Furthermore, r-SP exhibited potent activation of Toll-like receptor 2 compared with h-SP or f-SP. The expression of IL-6 and IL-8 induced by r-SP was mediated through the activation of mitogen-activated protein kinases. Remarkably, when r-SP was further treated with heat or formalin, there was a decrease in the aforementioned activities. Taken together, we suggest that r-SP stimulates the human respiratory epithelial cells to produce the cytokines IL-6 and IL-8, which might influence the induction of adaptive immune responses.
Collapse
Affiliation(s)
- Min Yong Jwa
- Department of Oral Microbiology and Immunology, DRI and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Eun Byeol Ko
- Department of Oral Microbiology and Immunology, DRI and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyun Young Kim
- Department of Oral Microbiology and Immunology, DRI and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sun Kyung Kim
- Department of Oral Microbiology and Immunology, DRI and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Soyoung Jeong
- Department of Oral Microbiology and Immunology, DRI and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ho Seong Seo
- Research Division for Biotechnology, Korea Atomic Energy Research Institute, Jeongeup, 56212, Republic of Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, DRI and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
16
|
Brooks LRK, Mias GI. Streptococcus pneumoniae's Virulence and Host Immunity: Aging, Diagnostics, and Prevention. Front Immunol 2018; 9:1366. [PMID: 29988379 PMCID: PMC6023974 DOI: 10.3389/fimmu.2018.01366] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/01/2018] [Indexed: 12/14/2022] Open
Abstract
Streptococcus pneumoniae is an infectious pathogen responsible for millions of deaths worldwide. Diseases caused by this bacterium are classified as pneumococcal diseases. This pathogen colonizes the nasopharynx of its host asymptomatically, but overtime can migrate to sterile tissues and organs and cause infections. Pneumonia is currently the most common pneumococcal disease. Pneumococcal pneumonia is a global health concern and vastly affects children under the age of five as well as the elderly and individuals with pre-existing health conditions. S. pneumoniae has a large selection of virulence factors that promote adherence, invasion of host tissues, and allows it to escape host immune defenses. A clear understanding of S. pneumoniae's virulence factors, host immune responses, and examining the current techniques available for diagnosis, treatment, and disease prevention will allow for better regulation of the pathogen and its diseases. In terms of disease prevention, other considerations must include the effects of age on responses to vaccines and vaccine efficacy. Ongoing work aims to improve on current vaccination paradigms by including the use of serotype-independent vaccines, such as protein and whole cell vaccines. Extending our knowledge of the biology of, and associated host immune response to S. pneumoniae is paramount for our improvement of pneumococcal disease diagnosis, treatment, and improvement of patient outlook.
Collapse
Affiliation(s)
- Lavida R. K. Brooks
- Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
- Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI, United States
| | - George I. Mias
- Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI, United States
- Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
17
|
Santos G, Lai X, Eberhardt M, Vera J. Bacterial Adherence and Dwelling Probability: Two Drivers of Early Alveolar Infection by Streptococcus pneumoniae Identified in Multi-Level Mathematical Modeling. Front Cell Infect Microbiol 2018; 8:159. [PMID: 29868515 PMCID: PMC5962665 DOI: 10.3389/fcimb.2018.00159] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 04/25/2018] [Indexed: 01/31/2023] Open
Abstract
Pneumococcal infection is the most frequent cause of pneumonia, and one of the most prevalent diseases worldwide. The population groups at high risk of death from bacterial pneumonia are infants, elderly and immunosuppressed people. These groups are more vulnerable because they have immature or impaired immune systems, the efficacy of their response to vaccines is lower, and antibiotic treatment often does not take place until the inflammatory response triggered is already overwhelming. The immune response to bacterial lung infections involves dynamic interactions between several types of cells whose activation is driven by intracellular molecular networks. A feasible approach to the integration of knowledge and data linking tissue, cellular and intracellular events and the construction of hypotheses in this area is the use of mathematical modeling. For this paper, we used a multi-level computational model to analyse the role of cellular and molecular interactions during the first 10 h after alveolar invasion of Streptococcus pneumoniae bacteria. By “multi-level” we mean that we simulated the interplay between different temporal and spatial scales in a single computational model. In this instance, we included the intracellular scale of processes driving lung epithelial cell activation together with the scale of cell-to-cell interactions at the alveolar tissue. In our analysis, we combined systematic model simulations with logistic regression analysis and decision trees to find genotypic-phenotypic signatures that explain differences in bacteria strain infectivity. According to our simulations, pneumococci benefit from a high dwelling probability and a high proliferation rate during the first stages of infection. In addition to this, the model predicts that during the very early phases of infection the bacterial capsule could be an impediment to the establishment of the alveolar infection because it impairs bacterial colonization.
Collapse
Affiliation(s)
- Guido Santos
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Universitätsklinikum Erlangen and Faculty of Medicine, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Xin Lai
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Universitätsklinikum Erlangen and Faculty of Medicine, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Eberhardt
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Universitätsklinikum Erlangen and Faculty of Medicine, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Julio Vera
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Universitätsklinikum Erlangen and Faculty of Medicine, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
18
|
Zhi X, Abdullah IT, Gazioglu O, Manzoor I, Shafeeq S, Kuipers OP, Hiller NL, Andrew PW, Yesilkaya H. Rgg-Shp regulators are important for pneumococcal colonization and invasion through their effect on mannose utilization and capsule synthesis. Sci Rep 2018; 8:6369. [PMID: 29686372 PMCID: PMC5913232 DOI: 10.1038/s41598-018-24910-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 04/05/2018] [Indexed: 01/18/2023] Open
Abstract
Microbes communicate with each other by using quorum sensing (QS) systems and modulate their collective 'behavior' for in-host colonization and virulence, biofilm formation, and environmental adaptation. The recent increase in genome data availability reveals the presence of several putative QS sensing circuits in microbial pathogens, but many of these have not been functionally characterized yet, despite their possible utility as drug targets. To increase the repertoire of functionally characterized QS systems in bacteria, we studied Rgg144/Shp144 and Rgg939/Shp939, two putative QS systems in the important human pathogen Streptococcus pneumoniae. We find that both of these QS circuits are induced by short hydrophobic peptides (Shp) upon sensing sugars found in the respiratory tract, such as galactose and mannose. Microarray analyses using cultures grown on mannose and galactose revealed that the expression of a large number of genes is controlled by these QS systems, especially those encoding for essential physiological functions and virulence-related genes such as the capsular locus. Moreover, the array data revealed evidence for cross-talk between these systems. Finally, these Rgg systems play a key role in colonization and virulence, as deletion mutants of these QS systems are attenuated in the mouse models of colonization and pneumonia.
Collapse
Affiliation(s)
- Xiangyun Zhi
- Department of Infection, Immunity & Inflammation, University of Leicester, Leicester, LE1 9HN, UK
| | - Iman Tajer Abdullah
- Department of Infection, Immunity & Inflammation, University of Leicester, Leicester, LE1 9HN, UK
- Department of Biology, College of Science, University of Kirkuk, Kirkuk, Iraq
| | - Ozcan Gazioglu
- Department of Infection, Immunity & Inflammation, University of Leicester, Leicester, LE1 9HN, UK
| | - Irfan Manzoor
- Molecular Genetics, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Sulman Shafeeq
- Molecular Genetics, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Oscar P Kuipers
- Molecular Genetics, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - N Luisa Hiller
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA, 15213, USA
| | - Peter W Andrew
- Department of Infection, Immunity & Inflammation, University of Leicester, Leicester, LE1 9HN, UK
| | - Hasan Yesilkaya
- Department of Infection, Immunity & Inflammation, University of Leicester, Leicester, LE1 9HN, UK.
| |
Collapse
|
19
|
Kozlowski E, Wasserman GA, Morgan M, O’Carroll D, Ramirez NGP, Gummuluru S, Rah JY, Gower AC, Ieong M, Quinton LJ, Mizgerd JP, Jones MR. The RNA uridyltransferase Zcchc6 is expressed in macrophages and impacts innate immune responses. PLoS One 2017; 12:e0179797. [PMID: 28665939 PMCID: PMC5493306 DOI: 10.1371/journal.pone.0179797] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 06/05/2017] [Indexed: 02/03/2023] Open
Abstract
Alveolar macrophages orchestrate pulmonary innate immunity and are essential for early immune surveillance and clearance of microorganisms in the airways. Inflammatory signaling must be sufficiently robust to promote host defense but limited enough to prevent excessive tissue injury. Macrophages in the lungs utilize multiple transcriptional and post-transcriptional mechanisms of inflammatory gene expression to delicately balance the elaboration of immune mediators. RNA terminal uridyltransferases (TUTs), including the closely homologous family members Zcchc6 (TUT7) and Zcchc11 (TUT4), have been implicated in the post-transcriptional regulation of inflammation from studies conducted in vitro. In vivo, we observed that Zcchc6 is expressed in mouse and human primary macrophages. Zcchc6-deficient mice are viable and born in Mendelian ratios and do not exhibit an observable spontaneous phenotype under basal conditions. Following an intratracheal challenge with S. pneumoniae, Zcchc6 deficiency led to a modest but significant increase in the expression of select cytokines including IL-6, CXCL1, and CXCL5. These findings were recapitulated in vitro whereby Zcchc6-deficient macrophages exhibited similar increases in cytokine expression due to bacterial stimulation. Although loss of Zcchc6 also led to increased neutrophil emigration to the airways during pneumonia, these responses were not sufficient to impact host defense against infection.
Collapse
Affiliation(s)
- Elyse Kozlowski
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Gregory A. Wasserman
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Marcos Morgan
- European Molecular Biology Laboratory (EMBL), Mouse Biology Unit, Monterotondo, Italy
| | - Dónal O’Carroll
- European Molecular Biology Laboratory (EMBL), Mouse Biology Unit, Monterotondo, Italy
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Nora-Guadalupe P. Ramirez
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Suryaram Gummuluru
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Jasmine Y. Rah
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Adam C. Gower
- Clinical and Translational Science Institute, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Michael Ieong
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Lee J. Quinton
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Joseph P. Mizgerd
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Matthew R. Jones
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
20
|
Gowing SD, Chow SC, Cools-Lartigue JJ, Chen CB, Najmeh S, Jiang HY, Bourdeau F, Beauchamp A, Mancini U, Angers I, Giannias B, Spicer JD, Rousseau S, Qureshi ST, Ferri LE. Gram-positive pneumonia augments non-small cell lung cancer metastasis via host toll-like receptor 2 activation. Int J Cancer 2017; 141:561-571. [PMID: 28401532 DOI: 10.1002/ijc.30734] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 03/23/2017] [Indexed: 12/29/2022]
Abstract
Surgical resection of early stage nonsmall cell lung cancer (NSCLC) is necessary for cure. However, rates of postoperative bacterial pneumonias remain high and may confer an increased risk for metastasis. Toll-like receptors (TLRs) mediate the inflammatory cascade by recognizing microbial products at the surface of numerous cell types in the lung; however, little is known about how host TLRs influence NSCLC metastasis. TLR2 recognizes gram-positive bacterial cell wall components activating innate immunity. We demonstrate that lower respiratory tract infection with Streptococcus pneumonia augments the formation of murine H59 NSCLC liver metastases in C57BL/6 mice through host TLR2 activation. Infected mice demonstrate increased H59 and human A549 NSCLC adhesion to hepatic sinusoids in vivo compared with noninfected controls, a response that is significantly diminished in TLR2 knock-out mice. Intra-tracheal injection of purified TLR2 ligand lipoteichoic acid into mice similarly augments in vivo adhesion of H59 cells to hepatic sinusoids. Additionally, H59 and A549 NSCLC cells incubated with bronchoepithelial conditioned media show increased cell adhesion to extracellular matrix components in vitro and hepatic sinusoids in vivo in a manner that is dependent on bronchoepithelial TLR2 activation and interleukin-6 secretion. TLR2 is therefore a potential therapeutic target for gram-positive pneumonia-driven NSCLC metastasis.
Collapse
Affiliation(s)
- Stephen D Gowing
- Deparment of Surgery, L.D. MacLean Surgical Research Laboratories, McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Simon C Chow
- Deparment of Surgery, L.D. MacLean Surgical Research Laboratories, McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Jonathan J Cools-Lartigue
- Deparment of Surgery, L.D. MacLean Surgical Research Laboratories, McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Crystal B Chen
- Deparment of Surgery, L.D. MacLean Surgical Research Laboratories, McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Sara Najmeh
- Deparment of Surgery, L.D. MacLean Surgical Research Laboratories, McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Henry Y Jiang
- Deparment of Surgery, L.D. MacLean Surgical Research Laboratories, McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - France Bourdeau
- Deparment of Surgery, L.D. MacLean Surgical Research Laboratories, McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Annie Beauchamp
- Department of Critical Care and Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Ugo Mancini
- Deparment of Surgery, L.D. MacLean Surgical Research Laboratories, McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Isabelle Angers
- Department of Critical Care and Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Betty Giannias
- Deparment of Surgery, L.D. MacLean Surgical Research Laboratories, McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Jonathan D Spicer
- Deparment of Surgery, L.D. MacLean Surgical Research Laboratories, McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Simon Rousseau
- Department of Medicine, Meakins-Christie Laboratories, McGill University Health Center, McGill University, Montreal, QC, Canada
| | - Salman T Qureshi
- Department of Critical Care and Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Lorenzo E Ferri
- Deparment of Surgery, L.D. MacLean Surgical Research Laboratories, McGill University Health Centre, McGill University, Montreal, QC, Canada
| |
Collapse
|
21
|
Turner AG, Ong CLY, Walker MJ, Djoko KY, McEwan AG. Transition Metal Homeostasis in Streptococcus pyogenes and Streptococcus pneumoniae. Adv Microb Physiol 2017; 70:123-191. [PMID: 28528647 DOI: 10.1016/bs.ampbs.2017.01.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Trace metals such as Fe, Mn, Zn and Cu are essential for various biological functions including proper innate immune function. The host immune system has complicated and coordinated mechanisms in place to either starve and/or overload invading pathogens with various metals to combat the infection. Here, we discuss the roles of Fe, Mn and Zn in terms of nutritional immunity, and also the roles of Cu and Zn in metal overload in relation to the physiology and pathogenesis of two human streptococcal species, Streptococcus pneumoniae and Streptococcus pyogenes. S. pneumoniae is a major human pathogen that is carried asymptomatically in the nasopharynx by up to 70% of the population; however, transition to internal sites can cause a range of diseases such as pneumonia, otitis media, meningitis and bacteraemia. S. pyogenes is a human pathogen responsible for diseases ranging from pharyngitis and impetigo, to severe invasive infections. Both species have overlapping capacity with respect to metal acquisition, export and regulation and how metal homeostasis relates to their virulence and ability to invade and survive within the host. It is becoming more apparent that metals have an important role to play in the control of infection, and with further investigations, it could lead to the potential use of metals in novel antimicrobial therapies.
Collapse
Affiliation(s)
- Andrew G Turner
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Cheryl-Lynn Y Ong
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Mark J Walker
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Karrera Y Djoko
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Alastair G McEwan
- School of Chemistry and Molecular Biosciences and Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
22
|
Mracsko E, Stegemann-Koniszewski S, Na SY, Dalpke A, Bruder D, Lasitschka F, Veltkamp R. A Mouse Model of Post-Stroke Pneumonia Induced by Intra-Tracheal Inoculation with Streptococcus pneumoniae. Cerebrovasc Dis 2017; 43:99-109. [DOI: 10.1159/000452136] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 09/22/2016] [Indexed: 02/04/2023] Open
Abstract
Background: Stroke-induced immunodeficiency increases the risk of infectious complications, which adversely affects neurological outcome. Among those, pneumonia affects as many as one third of stroke patients and is the main contributor to mortality in the post-acute phase of stroke. Experimental findings on post-stroke susceptibility to spontaneous pneumonia in mice are contradictory. Here, we established a mouse model inducing standardized bacterial pneumonia and characterized the impaired pulmonary cellular and humoral immune responses after experimental stroke. Methods: Bacterial pneumonia was induced by intra-tracheal inoculation with Streptococcus pneumoniae at different time points after transient middle cerebral artery occlusion (MCAO). Bacterial counts in lungs and blood, histological changes, and cytokine production in the lungs were assessed. Furthermore, we investigated the effect of pneumonia on stroke outcome. Results: Intra-tracheal inoculation resulted in reproducible pneumonia and bacteraemia, and demonstrated post-stroke susceptibility to streptococcal pneumonia developing with a delay of at least 24 h after MCAO. Higher bacterial counts in mice infected 3 days after stroke induction correlated with reduced neutrophil and macrophage infiltration in the lungs and lower levels of pro-inflammatory cytokines in the broncho-alveolar lavage compared to sham-operated animals. Pneumonia increased mortality without affecting brain-infiltrating leukocytes. Conclusions: In this standardized mouse model of post-stroke pneumonia, we describe attenuated leukocyte infiltration and cytokine production in response to bacterial infection in the lungs that has a profound effect on outcome.
Collapse
|
23
|
Streptococcus pneumoniae disrupts pulmonary immune defence via elastase release following pneumolysin-dependent neutrophil lysis. Sci Rep 2016; 6:38013. [PMID: 27892542 PMCID: PMC5125098 DOI: 10.1038/srep38013] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 11/04/2016] [Indexed: 11/08/2022] Open
Abstract
Streptococcus pneumoniae is a leading cause of bacterial pneumonia and is the principal cause of morbidity and mortality worldwide. Previous studies suggested that excessive activation of neutrophils results in the release of neutrophil elastase, which contributes to lung injury in severe pneumonia. Although both pneumococcal virulence factors and neutrophil elastase contribute to the development and progression of pneumonia, there are no studies analysing relationships between these factors. Here, we showed that pneumolysin, a pneumococcal pore-forming toxin, induced cell lysis in primary isolated human neutrophils, leading to the release of neutrophil elastase. Pneumolysin exerted minimal cytotoxicity against alveolar epithelial cells and macrophages, whereas neutrophil elastase induced detachment of alveolar epithelial cells and impaired phagocytic activity in macrophages. Additionally, activation of neutrophil elastase did not exert bactericidal activity against S. pneumoniae in vitro. P2X7 receptor, which belongs to a family of purinergic receptors, was involved in pneumolysin-induced cell lysis. These findings suggested that infiltrated neutrophils are the primary target cells of pneumolysin, and that S. pneumoniae exploits neutrophil-elastase leakage to induce the disruption of pulmonary immune defences, thereby causing lung injury.
Collapse
|
24
|
Lung epithelium and myeloid cells cooperate to clear acute pneumococcal infection. Mucosal Immunol 2016; 9:1288-302. [PMID: 26627460 PMCID: PMC4990776 DOI: 10.1038/mi.2015.128] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 11/01/2015] [Indexed: 02/04/2023]
Abstract
The Gram-positive bacterium Streptococcus pneumoniae causes life-threatening infections, especially among immunocompromised patients. The host's immune system senses S. pneumoniae via different families of pattern recognition receptors, in particular the Toll-like receptor (TLR) family that promotes immune cell activation. Yet, while single TLRs are dispensable for initiating inflammatory responses against S. pneumoniae, the central TLR adapter protein myeloid differentiation factor 88 (MyD88) is of vital importance, as MyD88-deficient mice succumb rapidly to infection. Since MyD88 is ubiquitously expressed in hematopoietic and non-hematopoietic cells, the extent to which MyD88 signaling is required in different cell types to control S. pneumoniae is unknown. Therefore, we used novel conditional knockin mice to investigate the necessity of MyD88 signaling in distinct lung-resident myeloid and epithelial cells for the initiation of a protective immune response against S. pneumoniae. Here, we show that MyD88 signaling in lysozyme M (LysM)- and CD11c-expressing myeloid cells, as well as in pulmonary epithelial cells, is critical to restore inflammatory cytokine and antimicrobial peptide production, leading to efficient neutrophil recruitment and enhanced bacterial clearance. Overall, we show a novel synergistic requirement of compartment-specific MyD88 signaling in S. pneumoniae immunity.
Collapse
|
25
|
Jim KK, Engelen-Lee J, van der Sar AM, Bitter W, Brouwer MC, van der Ende A, Veening JW, van de Beek D, Vandenbroucke-Grauls CMJE. Infection of zebrafish embryos with live fluorescent Streptococcus pneumoniae as a real-time pneumococcal meningitis model. J Neuroinflammation 2016; 13:188. [PMID: 27542968 PMCID: PMC4992281 DOI: 10.1186/s12974-016-0655-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 07/08/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Streptococcus pneumoniae is one of the most important causes of bacterial meningitis, an infection where unfavourable outcome is driven by bacterial and host-derived toxins. In this study, we developed and characterized a pneumococcal meningitis model in zebrafish embryos that allows for real-time investigation of early host-microbe interaction. METHODS Zebrafish embryos were infected in the caudal vein or hindbrain ventricle with green fluorescent wild-type S. pneumoniae D39 or a pneumolysin-deficient mutant. The kdrl:mCherry transgenic zebrafish line was used to visualize the blood vessels, whereas phagocytic cells were visualized by staining with far red anti-L-plastin or in mpx:GFP/mpeg1:mCherry zebrafish, that have green fluorescent neutrophils and red fluorescent macrophages. Imaging was performed by fluorescence confocal and time-lapse microscopy. RESULTS After infection by caudal vein, we saw focal clogging of the pneumococci in the blood vessels and migration of bacteria through the blood-brain barrier into the subarachnoid space and brain tissue. Infection with pneumolysin-deficient S. pneumoniae in the hindbrain ventricle showed attenuated growth and migration through the brain as compared to the wild-type strain. Time-lapse and confocal imaging revealed that the initial innate immune response to S. pneumoniae in the subarachnoid space mainly consisted of neutrophils and that pneumolysin-mediated cytolytic activity caused a marked reduction of phagocytes. CONCLUSIONS This new meningitis model permits detailed analysis and visualization of host-microbe interaction in pneumococcal meningitis in real time and is a very promising tool to further our insights in the pathogenesis of pneumococcal meningitis.
Collapse
Affiliation(s)
- Kin Ki Jim
- Department of Medical Microbiology and Infection Control, VU University Medical Center, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
- Department of Neurology, Center of Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - JooYeon Engelen-Lee
- Department of Neurology, Center of Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Astrid M van der Sar
- Department of Medical Microbiology and Infection Control, VU University Medical Center, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Wilbert Bitter
- Department of Medical Microbiology and Infection Control, VU University Medical Center, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Matthijs C Brouwer
- Department of Neurology, Center of Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Arie van der Ende
- Department of Medical Microbiology, Center of Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- The Netherlands Reference Laboratory for Bacterial Meningitis, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Jan-Willem Veening
- Molecular Genetics Group, Groningen Biomolecular Sciences and Biotechnology Institute, Centre for Synthetic Biology, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Diederik van de Beek
- Department of Neurology, Center of Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Christina M J E Vandenbroucke-Grauls
- Department of Medical Microbiology and Infection Control, VU University Medical Center, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands.
- Department of Medical Microbiology and Infection Control, VU University Medical Center, P.O. Box 7057, 1007 MB, Amsterdam, The Netherlands.
| |
Collapse
|
26
|
Abstract
Streptococcus pneumoniae is an opportunistic pathogen globally associated with significant morbidity and mortality. It is capable of causing a wide range of diseases including sinusitis, conjunctivitis, otitis media, pneumonia, bacteraemia, sepsis, and meningitis. While its capsular polysaccharide is indispensible for invasive disease, and opsonising antibodies against the capsule are the basis for the current vaccines, a long history of biomedical research indicates that other components of this Gram-positive bacterium are also critical for virulence. Herein we review the contribution of pneumococcal virulence determinants to survival and persistence in the context of distinct anatomical sites. We discuss how these determinants allow the pneumococcus to evade mucociliary clearance during colonisation, establish lower respiratory tract infection, resist complement deposition and opsonophagocytosis in the bloodstream, and invade secondary tissues such as the central nervous system leading to meningitis. We do so in a manner that highlights both the critical role of the capsular polysaccharide and the accompanying and necessary protein determinants. Understanding the complex interplay between host and pathogen is necessary to find new ways to prevent pneumococcal infection. This review is an attempt to do so with consideration for the latest research findings.
Collapse
|
27
|
Nel JG, Theron AJ, Durandt C, Tintinger GR, Pool R, Mitchell TJ, Feldman C, Anderson R. Pneumolysin activates neutrophil extracellular trap formation. Clin Exp Immunol 2016; 184:358-67. [PMID: 26749379 DOI: 10.1111/cei.12766] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 12/14/2015] [Accepted: 12/23/2015] [Indexed: 02/06/2023] Open
Abstract
The primary objective of the current study was to investigate the potential of the pneumococcal toxin, pneumolysin (Ply), to activate neutrophil extracellular trap (NET) formation in vitro. Isolated human blood neutrophils were exposed to recombinant Ply (5-20 ng ml(-1) ) for 30-90 min at 37°C and NET formation measured using the following procedures to detect extracellular DNA: (i) flow cytometry using Vybrant® DyeCycle™ Ruby; (ii) spectrofluorimetry using the fluorophore, Sytox(®) Orange (5 μM); and (iii) NanoDrop(®) technology. These procedures were complemented by fluorescence microscopy using 4', 6-diamino-2-phenylindole (DAPI) (nuclear stain) in combination with anti-citrullinated histone monoclonal antibodies to visualize nets. Exposure of neutrophils to Ply resulted in relatively rapid (detected within 30-60 min), statistically significant (P < 0·05) dose- and time-related increases in the release of cellular DNA impregnated with both citrullinated histone and myeloperoxidase. Microscopy revealed that NETosis appeared to be restricted to a subpopulation of neutrophils, the numbers of NET-forming cells in the control and Ply-treated systems (10 and 20 ng ml(-1) ) were 4·3 (4·2), 14.3 (9·9) and 16·5 (7·5), respectively (n = 4, P < 0·0001 for comparison of the control with both Ply-treated systems). Ply-induced NETosis occurred in the setting of retention of cell viability, and apparent lack of involvement of reactive oxygen species and Toll-like receptor 4. In conclusion, Ply induces vital NETosis in human neutrophils, a process which may either contribute to host defence or worsen disease severity, depending on the intensity of the inflammatory response during pneumococcal infection.
Collapse
Affiliation(s)
| | - A J Theron
- Department of Immunology, Faculty of Health Sciences, University of Pretoria and Tshwane Academic Division of the National Health Laboratory Service.,South African Medical Research Council Unit for Stem Cell Research, Institute for Cellular and Molecular Medicine, Department of Immunology, Faculty of Health Sciences, University of Pretoria
| | - C Durandt
- South African Medical Research Council Unit for Stem Cell Research, Institute for Cellular and Molecular Medicine, Department of Immunology, Faculty of Health Sciences, University of Pretoria
| | - G R Tintinger
- Department of Internal Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - R Pool
- Department of Haematology
| | - T J Mitchell
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - C Feldman
- Division of Pulmonology, Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg and Charlotte Maxeke Academic Hospital, Johannesburg, South Africa
| | - R Anderson
- South African Medical Research Council Unit for Stem Cell Research, Institute for Cellular and Molecular Medicine, Department of Immunology, Faculty of Health Sciences, University of Pretoria
| |
Collapse
|
28
|
Hommes TJ, van Lieshout MH, van ‘t Veer C, Florquin S, Bootsma HJ, Hermans PW, de Vos AF, van der Poll T. Role of Nucleotide-Binding Oligomerization Domain-Containing (NOD) 2 in Host Defense during Pneumococcal Pneumonia. PLoS One 2015; 10:e0145138. [PMID: 26673231 PMCID: PMC4682639 DOI: 10.1371/journal.pone.0145138] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 11/26/2015] [Indexed: 11/18/2022] Open
Abstract
Streptococcus (S.) pneumoniae is the most common causative pathogen in community-acquired pneumonia. Nucleotide-binding oligomerization domain-containing (NOD) 2 is a pattern recognition receptor located in the cytosol of myeloid cells that is able to detect peptidoglycan fragments of S. pneumoniae. We here aimed to investigate the role of NOD2 in the host response during pneumococcal pneumonia. Phagocytosis of S. pneumoniae was studied in NOD2 deficient (Nod2-/-) and wild-type (Wt) alveolar macrophages and neutrophils in vitro. In subsequent in vivo experiments Nod2-/- and Wt mice were inoculated with serotype 2 S. pneumoniae (D39), an isogenic capsule locus deletion mutant (D39Δcps) or serotype 3 S. pneumoniae (6303) via the airways, and bacterial growth and dissemination and the lung inflammatory response were evaluated. Nod2-/- alveolar macrophages and blood neutrophils displayed a reduced capacity to internalize pneumococci in vitro. During pneumonia caused by S. pneumoniae D39 Nod2-/- mice were indistinguishable from Wt mice with regard to bacterial loads in lungs and distant organs, lung pathology and neutrophil recruitment. While Nod2-/- and Wt mice also had similar bacterial loads after infection with the more virulent S. pneumoniae 6303 strain, Nod2-/- mice displayed a reduced bacterial clearance of the normally avirulent unencapsulated D39Δcps strain. These results suggest that NOD2 does not contribute to host defense during pneumococcal pneumonia and that the pneumococcal capsule impairs recognition of S. pneumoniae by NOD2.
Collapse
Affiliation(s)
- Tijmen J. Hommes
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Center for Infection and Immunity, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- * E-mail:
| | - Miriam H. van Lieshout
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Center for Infection and Immunity, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Cornelis van ‘t Veer
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Center for Infection and Immunity, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Sandrine Florquin
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Hester J. Bootsma
- Laboratory of Pediatric Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Peter W. Hermans
- Laboratory of Pediatric Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Alex F. de Vos
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Center for Infection and Immunity, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Center for Infection and Immunity, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
29
|
Otero C, Díaz D, Uriarte I, Bezrodnik L, Finiasz MR, Fink S. Peripheral blood monocyte and T cell subsets in children with specific polysaccharide antibody deficiency (SPAD). Hum Immunol 2015; 77:12-19. [PMID: 26577026 DOI: 10.1016/j.humimm.2015.09.053] [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: 02/10/2014] [Revised: 11/05/2014] [Accepted: 09/28/2015] [Indexed: 11/16/2022]
Abstract
Specific polysaccharide antibody deficiency (SPAD) is a well reported immunodeficiency characterized by a failure to produce antibodies against polyvalent polysaccharide antigens, expressed by encapsulated microorganisms. The clinical presentation of these patients involves recurrent bacterial infections, being the most frequent agent Streptococcus (S.) pneumoniae. In SPAD patients few reports refer to cells other than B cells. Since the immune response to S. pneumoniae and other encapsulated bacteria was historically considered restricted to B cells, the antibody deficiency seemed enough to justify the repetitive infections in SPAD patients. Our purpose is to determine if the B cell defects reported in SPAD patients are accompanied by defects in other leukocyte subpopulations necessary for the development of a proper adaptive immune response against S. pneumoniae. We here report that age related changes observed in healthy children involving increased percentages of classical monocytes (CD14++ CD16- cells) and decreased intermediate monocytes (CD14++ CD16+ cells), are absent in SPAD patients. Alterations can also be observed in T cells, supporting that the immune deficiency in SPAD patients is more complex than what has been described up to now.
Collapse
Affiliation(s)
- C Otero
- Immunology Department, IMEX-CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - D Díaz
- Immunology, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - I Uriarte
- Immunology, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - L Bezrodnik
- Immunology, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - M R Finiasz
- Immunology Department, IMEX-CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - S Fink
- Immunology Department, IMEX-CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina.
| |
Collapse
|
30
|
Hergott CB, Roche AM, Naidu NA, Mesaros C, Blair IA, Weiser JN. Bacterial exploitation of phosphorylcholine mimicry suppresses inflammation to promote airway infection. J Clin Invest 2015; 125:3878-90. [PMID: 26426079 DOI: 10.1172/jci81888] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 07/23/2015] [Indexed: 12/22/2022] Open
Abstract
Regulation of neutrophil activity is critical for immune evasion among extracellular pathogens, yet the mechanisms by which many bacteria disrupt phagocyte function remain unclear. Here, we have shown that the respiratory pathogen Streptococcus pneumoniae disables neutrophils by exploiting molecular mimicry to degrade platelet-activating factor (PAF), a host-derived inflammatory phospholipid. Using mass spectrometry and murine upper airway infection models, we demonstrated that phosphorylcholine (ChoP) moieties that are shared by PAF and the bacterial cell wall allow S. pneumoniae to leverage a ChoP-remodeling enzyme (Pce) to remove PAF from the airway. S. pneumoniae-mediated PAF deprivation impaired viability, activation, and bactericidal capacity among responding neutrophils. In the absence of Pce, neutrophils rapidly cleared S. pneumoniae from the airway and impeded invasive disease and transmission between mice. Abrogation of PAF signaling rendered Pce dispensable for S. pneumoniae persistence, reinforcing that this enzyme deprives neutrophils of essential PAF-mediated stimulation. Accordingly, exogenous activation of neutrophils overwhelmed Pce-mediated phagocyte disruption. Haemophilus influenzae also uses an enzyme, GlpQ, to hydrolyze ChoP and subvert PAF function, suggesting that mimicry-driven immune evasion is a common paradigm among respiratory pathogens. These results identify a mechanism by which shared molecular structures enable microbial enzymes to subvert host lipid signaling, suppress inflammation, and ensure bacterial persistence at the mucosa.
Collapse
|
31
|
Tumor necrosis factor-alpha deficiency impairs host defense against Streptococcus pneumoniae. Lab Anim Res 2015; 31:78-85. [PMID: 26155202 PMCID: PMC4490149 DOI: 10.5625/lar.2015.31.2.78] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 05/21/2015] [Accepted: 05/31/2015] [Indexed: 11/25/2022] Open
Abstract
Streptococcus pneumoniae is a major human pathogen that is involved in community-acquired pneumonia. Tumor necrosis factor-alpha (TNF-α) is a pro-inflammatory cytokine that activates immune responses against infection, invasion, injury, or inflammation. To study the role of TNF-α during S. pneumoniae infection, a murine pneumococcal pneumonia model was used. We intranasally infected C57BL/6J wild-type (WT) and TNF-α knockout (KO) mice with S. pneumoniae D39 serotype 2. In TNF-α KO mice, continuous and distinct loss of body weight, and low survival rates were observed. Bacterial counts in the lungs and blood of TNF-α KO mice were significantly higher than those in WT mice. Histopathological lesions in the spleen of TNF-α KO mice were more severe than those in WT mice. In TNF-α KO mice, severe depletion of white pulp was observed and the number of apoptotic cells was significantly increased. Interferon-gamma (IFN-γ), IL-12p70 and IL-10 levels in serum were significantly increased in TNF-α KO mice. TNF-α is clearly involved in the regulation of S. pneumoniae infections. Early death and low survival rates of TNF-α KO mice were likely caused by a combination of impaired bacterial clearance and damage to the spleen. Our findings suggest that TNF-α plays a critical role in protecting the host from systemic S. pneumoniae infection.
Collapse
|
32
|
Han L, Kitova EN, Li J, Nikjah S, Lin H, Pluvinage B, Boraston AB, Klassen JS. Protein–Glycolipid Interactions Studied in Vitro Using ESI-MS and Nanodiscs: Insights into the Mechanisms and Energetics of Binding. Anal Chem 2015; 87:4888-96. [DOI: 10.1021/acs.analchem.5b00678] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Ling Han
- Alberta
Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - Elena N. Kitova
- Alberta
Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - Jun Li
- Alberta
Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - Sanaz Nikjah
- Alberta
Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - Hong Lin
- Alberta
Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - Benjamin Pluvinage
- Department
of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada V8W 3P6
| | - Alisdair B. Boraston
- Department
of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada V8W 3P6
| | - John S. Klassen
- Alberta
Glycomics Centre and Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| |
Collapse
|
33
|
The polysaccharide capsule of Streptococcus pneumonia partially impedes MyD88-mediated immunity during pneumonia in mice. PLoS One 2015; 10:e0118181. [PMID: 25700108 PMCID: PMC4336322 DOI: 10.1371/journal.pone.0118181] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 01/06/2015] [Indexed: 12/11/2022] Open
Abstract
Toll-like receptors (TLR) and the downstream adaptor protein MyD88 are considered crucial for protective immunity during bacterial infections. Streptococcus (S.) pneumoniae is a human respiratory pathogen and a large majority of clinical pneumococcal isolates expresses an external polysaccharide capsule. We here sought to determine the role of pneumococcal capsule in MyD88-mediated antibacterial defense during S. pneumonia pneumonia. Wild type (WT) and Myd88-/- mice were inoculated intranasally with serotype 2 S. pneumoniae D39 or with an isogenic capsule locus deletion mutant (D39∆cps), and analysed for bacterial outgrowth and inflammatory responses in the lung. As compared to WT mice, Myd88-/- mice infected with D39 demonstrated a modestly impaired bacterial clearance accompanied by decreased inflammatory responses in the lung. Strikingly, while WT mice rapidly cleared D39∆cps, Myd88-/- mice showed 105-fold higher bacterial burdens in their lungs and dissemination to blood 24 hours after infection. These data suggest that the pneumococcal capsule impairs recognition of TLR ligands expressed by S. pneumoniae and thereby partially impedes MyD88-mediated antibacterial defense.
Collapse
|
34
|
López Hernández Y, Yero D, Pinos-Rodríguez JM, Gibert I. Animals devoid of pulmonary system as infection models in the study of lung bacterial pathogens. Front Microbiol 2015; 6:38. [PMID: 25699030 PMCID: PMC4316775 DOI: 10.3389/fmicb.2015.00038] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 01/12/2015] [Indexed: 01/15/2023] Open
Abstract
Biological disease models can be difficult and costly to develop and use on a routine basis. Particularly, in vivo lung infection models performed to study lung pathologies use to be laborious, demand a great time and commonly are associated with ethical issues. When infections in experimental animals are used, they need to be refined, defined, and validated for their intended purpose. Therefore, alternative and easy to handle models of experimental infections are still needed to test the virulence of bacterial lung pathogens. Because non-mammalian models have less ethical and cost constraints as a subjects for experimentation, in some cases would be appropriated to include these models as valuable tools to explore host-pathogen interactions. Numerous scientific data have been argued to the more extensive use of several kinds of alternative models, such as, the vertebrate zebrafish (Danio rerio), and non-vertebrate insects and nematodes (e.g., Caenorhabditis elegans) in the study of diverse infectious agents that affect humans. Here, we review the use of these vertebrate and non-vertebrate models in the study of bacterial agents, which are considered the principal causes of lung injury. Curiously none of these animals have a respiratory system as in air-breathing vertebrates, where respiration takes place in lungs. Despite this fact, with the present review we sought to provide elements in favor of the use of these alternative animal models of infection to reveal the molecular signatures of host-pathogen interactions.
Collapse
Affiliation(s)
- Yamilé López Hernández
- Centro de Biociencias, Universidad Autónoma de San Luis Potosí San Luis de Potosí, Mexico
| | - Daniel Yero
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona Barcelona, Spain ; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona Barcelona, Spain
| | - Juan M Pinos-Rodríguez
- Centro de Biociencias, Universidad Autónoma de San Luis Potosí San Luis de Potosí, Mexico
| | - Isidre Gibert
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona Barcelona, Spain ; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona Barcelona, Spain
| |
Collapse
|
35
|
Williams AE, José RJ, Brown JS, Chambers RC. Enhanced inflammation in aged mice following infection with Streptococcus pneumoniae is associated with decreased IL-10 and augmented chemokine production. Am J Physiol Lung Cell Mol Physiol 2015; 308:L539-49. [PMID: 25595646 PMCID: PMC4360060 DOI: 10.1152/ajplung.00141.2014] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Streptococcus pneumoniae is the most common cause of severe pneumonia in the elderly. However, the impact of aging on the innate inflammatory response to pneumococci is poorly defined. We compared the innate immune response in old vs. young adult mice following infection with S. pneumoniae. The accumulation of neutrophils recovered from bronchoalveolar lavage fluid and lung homogenates was increased in aged compared with young adult mice, although bacterial outgrowth was similar in both age groups, as were markers of microvascular leak. Aged mice had similar levels of IL-1β, TNF, IFN-γ, IL-17, and granulocyte colony-stimulating factor following S. pneumoniae infection, compared with young mice, but increased levels of the chemokines CXCL9, CXCL12, CCL3, CCL4, CCL5, CCL11, and CCL17. Moreover, levels of IL-10 were significantly lower in aged animals. Neutralization of IL-10 in infected young mice was associated with increased neutrophil recruitment but no decrease in bacterial outgrowth. Furthermore, IL-10 neutralization resulted in increased levels of CCL3, CCL5, and CXCL10. We conclude that aging is associated with enhanced inflammatory responses following S. pneumoniae infection as a result of a compromised immunomodulatory cytokine response.
Collapse
Affiliation(s)
- Andrew E Williams
- Centre for Inflammation and Tissue Repair, Rayne Institute, University College London (UCL), London, United Kingdom
| | - Ricardo J José
- Centre for Inflammation and Tissue Repair, Rayne Institute, University College London (UCL), London, United Kingdom
| | - Jeremy S Brown
- Centre for Inflammation and Tissue Repair, Rayne Institute, University College London (UCL), London, United Kingdom
| | - Rachel C Chambers
- Centre for Inflammation and Tissue Repair, Rayne Institute, University College London (UCL), London, United Kingdom
| |
Collapse
|
36
|
Harvey RM, Hughes CE, Paton AW, Trappetti C, Tweten RK, Paton JC. The impact of pneumolysin on the macrophage response to Streptococcus pneumoniae is strain-dependent. PLoS One 2014; 9:e103625. [PMID: 25105894 PMCID: PMC4126675 DOI: 10.1371/journal.pone.0103625] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 06/28/2014] [Indexed: 11/18/2022] Open
Abstract
Streptococcus pneumoniae is the world's leading cause of pneumonia, bacteremia, meningitis and otitis media. A major pneumococcal virulence factor is the cholesterol-dependent cytolysin, which has the defining property of forming pores in cholesterol-containing membranes. In recent times a clinically significant and internationally successful serotype 1 ST306 clone has been found to express a non-cytolytic variant of Ply (Ply306). However, while the pneumococcus is a naturally transformable organism, strains of the ST306 clonal group have to date been virtually impossible to transform, severely restricting efforts to understand the role of non-cytolytic Ply in the success of this clone. In this study isogenic Ply mutants were constructed in the D39 background and for the first time in the ST306 background (A0229467) to enable direct comparisons between Ply variants for their impact on the immune response in a macrophage-like cell line. Strains that expressed cytolytic Ply were found to induce a significant increase in IL-1β release from macrophage-like cells compared to the non-cytolytic and Ply-deficient strains in a background-independent manner, confirming the requirement for pore formation in the Ply-dependent activation of the NLRP3 inflammasome. However, cytolytic activity in the D39 background was found to induce increased expression of the genes encoding GM-CSF (CSF2), p19 subunit of IL-23 (IL23A) and IFNβ (IFNB1) compared to non-cytolytic and Ply-deficient D39 mutants, but had no effect in the A0229467 background. The impact of Ply on the immune response to the pneumococcus is highly dependent on the strain background, thus emphasising the importance of the interaction between specific virulence factors and other components of the genetic background of this organism.
Collapse
Affiliation(s)
- Richard M. Harvey
- Research Centre for Infectious Diseases, School of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | - Catherine E. Hughes
- Research Centre for Infectious Diseases, School of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | - Adrienne W. Paton
- Research Centre for Infectious Diseases, School of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | - Claudia Trappetti
- Research Centre for Infectious Diseases, School of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | - Rodney K. Tweten
- Department of Microbiology & Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - James C. Paton
- Research Centre for Infectious Diseases, School of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
- * E-mail:
| |
Collapse
|
37
|
De Filippo K, Neill DR, Mathies M, Bangert M, McNeill E, Kadioglu A, Hogg N. A new protective role for S100A9 in regulation of neutrophil recruitment during invasive pneumococcal pneumonia. FASEB J 2014; 28:3600-8. [PMID: 24776746 DOI: 10.1096/fj.13-247460] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The S100A8/A9 heterodimer is abundantly expressed by myeloid cells, especially neutrophils, but its mechanism of action is only partially determined. In this study we investigated S100A8/A9 involvement in the host response to Streptococcus pneumoniae infection making use of S100a9(-/-) mice that lack heterodimer expression in myeloid cells. S100a9(-/-) mice that were infected intranasally with pneumococci rapidly succumbed, with 80% mortality after 48 h, whereas the majority of wild-type mice recovered. Over this time period, S100a9(-/-) mice displayed an average 6-fold reduction in circulating and lung-recruited neutrophils. Taqman analysis of S100a9(-/-) lungs revealed decreased production of a dominant subset of 5 cytokines and chemokines associated with neutrophil recruitment. The greatest differential was with the cytokine granulocyte colony-stimulating factor (G-CSF) that causes bone marrow release of neutrophils into the circulation (1900-fold difference at 48 h). Treating S100a9(-/-) mice with G-CSF reversed their increased susceptibility to infection by enhancing both circulating neutrophils and neutrophil recruitment into infected lungs, by reducing pneumococcal colony forming units, and by elevation of chemokine CXCL1, cytokine IL-6, and endogenous G-CSF proteins. Thus S100A9, potentially with its partner S100A8, makes a major contribution in the host response to pneumococcal infection by increasing circulating neutrophils principally regulation of G-CSF production.
Collapse
Affiliation(s)
- Katia De Filippo
- Leukocyte Adhesion Laboratory, Cancer Research United Kingdom London Research Institute, London, UK
| | - Daniel R Neill
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK; and Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, UK
| | - Meg Mathies
- Leukocyte Adhesion Laboratory, Cancer Research United Kingdom London Research Institute, London, UK
| | - Mathieu Bangert
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK; and Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, UK
| | - Eileen McNeill
- Leukocyte Adhesion Laboratory, Cancer Research United Kingdom London Research Institute, London, UK
| | - Aras Kadioglu
- Institute of Infection and Global Health, University of Liverpool, Liverpool, UK; and Department of Infection, Immunity, and Inflammation, University of Leicester, Leicester, UK
| | - Nancy Hogg
- Leukocyte Adhesion Laboratory, Cancer Research United Kingdom London Research Institute, London, UK;
| |
Collapse
|
38
|
Menter T, Giefing-Kroell C, Grubeck-Loebenstein B, Tzankov A. Characterization of the inflammatory infiltrate in Streptococcus pneumoniae pneumonia in young and elderly patients. Pathobiology 2014; 81:160-7. [PMID: 24751977 DOI: 10.1159/000360165] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Accepted: 01/27/2014] [Indexed: 11/19/2022] Open
Abstract
There is an increased susceptibility and mortality in the elderly due to pneumonia caused by Streptococcus pneumoniae. We aimed to assess the inflammatory cell composition with respect to age in pneumococcal pneumonia patients. Neutrophilic granulocytes and various lymphocyte and macrophage subpopulations were immunohistochemically quantified on lung tissue specimens of young (n = 5; mean age 8.4 years), middle-aged (n = 8; mean age 55.9 years) and elderly (n = 9; mean age 86.6 years) pneumonia patients with microbiologically proven S. pneumoniae pneumonia. We discovered a higher percentage of neutrophilic granulocytes in elderly as opposed to young patients (95 vs. 75%, p = 0.012). Conversely, young patients versus elderly patients had more alveolar macrophages (CD11c+: 20 vs. 9%, p = 0.029) and M1 macrophages (CD14+: 30 vs. 10%, p = 0.012 and HLA-DR+: 52 vs. 11%, p = 0.019). There was no significant difference concerning M2 macrophages and lymphocytes. Comparison of young patients with middle-aged patients showed similar significant results for alveolar macrophages (p = 0.019) and subsignificant results for M1 macrophages and neutrophilic granulocytes (p < 0.08). This is the first study characterizing the inflammatory infiltrate of pneumococcal pneumonia in situ. Our observations improve the understanding of the innate immune mechanisms of pneumococcal lung infection and point at the potential of therapies for restoring macrophage function and decreasing neutrophilic influx in order to help prevent or cure pneumonia.
Collapse
Affiliation(s)
- Thomas Menter
- Institute of Pathology, University Hospital, Basel, Switzerland
| | | | | | | |
Collapse
|
39
|
Dénes Á, Pradillo JM, Drake C, Sharp A, Warn P, Murray KN, Rohit B, Dockrell DH, Chamberlain J, Casbolt H, Francis S, Martinecz B, Nieswandt B, Rothwell NJ, Allan SM. Streptococcus pneumoniae worsens cerebral ischemia via interleukin 1 and platelet glycoprotein Ibα. Ann Neurol 2014; 75:670-83. [PMID: 24644058 DOI: 10.1002/ana.24146] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 03/17/2014] [Accepted: 03/17/2014] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Bacterial infection contributes to diverse noninfectious diseases and worsens outcome after stroke. Streptococcus pneumoniae, the most common infection in patients at risk of stroke, is a major cause of prolonged hospitalization and death of stroke patients, but how infection impacts clinical outcome is not known. METHODS We induced sustained pulmonary infection by a human S. pneumoniae isolate in naive and comorbid rodents to investigate the effect of infection on vascular and inflammatory responses prior to and after cerebral ischemia. RESULTS S. pneumoniae infection triggered atherogenesis, led to systemic induction of interleukin (IL) 1, and profoundly exacerbated (50-90%) ischemic brain injury in rats and mice, a response that was more severe in combination with old age and atherosclerosis. Systemic blockade of IL-1 with IL-1 receptor antagonist (IL-1Ra) fully reversed infection-induced exacerbation of brain injury and functional impairment caused by cerebral ischemia. We show that infection-induced systemic inflammation mediates its effects via increasing platelet activation and microvascular coagulation in the brain after cerebral ischemia, as confirmed by reduced brain injury in response to blockade of platelet glycoprotein (GP) Ibα. IL-1 and platelet-mediated signals converge on microglia, as both IL-1Ra and GPIbα blockade reversed the production of IL-1α by microglia in response to cerebral ischemia in infected animals. INTERPRETATION S. pneumoniae infection augments atherosclerosis and exacerbates ischemic brain injury via IL-1 and platelet-mediated systemic inflammation. These mechanisms may contribute to diverse cardio- and cerebrovascular pathologies in humans.
Collapse
Affiliation(s)
- Ádám Dénes
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom; Laboratory of Molecular Neuroendocrinology, Institute of Experimental Medicine, Budapest, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Karadeniz G, Polat G, Senol G, Buyuksirin M. C-reactive protein measurements as a marker of the severity of chronic obstructive pulmonary disease exacerbations. Inflammation 2014; 36:948-53. [PMID: 23547018 DOI: 10.1007/s10753-013-9625-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory disease of the lungs. Acute exacerbations of COPD (AE-COPD) are a result of infectious or non-infectious instances. In our study, we aimed to determine whether serum C-reactive protein (CRP) levels are predictive indicators for disease severity and prognosis in hospitalized patients with AE-COPD. A total of 64 patients (36 regular ward and 28 ICU patients) were included in the study. Cases were identified and classified according to the Global Initiative for COPD. The first CRP test levels at acceptance at the ward or intensive care unit were counted in the study. CRP levels of patients in intensive care were significantly higher than those of patients in the regular ward. Mean values of CRP were detected to be 6.28 ± 6.53 mg/dl in the regular ward cases and 16.9 ± 12.03 mg/dl in the ICU patients (p < 0.01). The stage of COPD did not indicate a significant difference in terms of CRP values. Mean CRP values were found to be 16.02 ± 6.95 mg/dl in mortal cases and 9.76 ± 11.09 mg/dl in survivors (p < 0.01). High CRP levels were considered as a prognostic parameter and indicator of severity of AE-COPD. Increased mortality risk was found to be associated with high CRP values.
Collapse
Affiliation(s)
- Gulistan Karadeniz
- Department of Chest Diseases, Izmir Chest Diseases and Thoracic Surgery Hospital, Izmir, Turkey
| | | | | | | |
Collapse
|
41
|
Damjanovic D, Khera A, Medina MF, Ennis J, Turner JD, Gauldie J, Xing Z. Type 1 interferon gene transfer enhances host defense against pulmonary Streptococcus pneumoniae infection via activating innate leukocytes. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2014; 1:5. [PMID: 26015944 PMCID: PMC4378291 DOI: 10.1038/mtm.2014.5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 01/29/2014] [Indexed: 11/11/2022]
Abstract
Pneumococcal infections are the leading cause of community-acquired pneumonia. Although the type 1 interferon-α (IFN-α) is a well-known antiviral cytokine, the role of IFN-α in antipneumococcal host defense and its therapeutic potential remain poorly understood. We have investigated these issues by using a murine transgene expression model. We found that in control animals, Streptococcus pneumoniae infection caused severe weight loss and excessive lung inflammation, associated with rapid bacterial outgrowth. In contrast, the animals that received a single dose of an adenoviral vector expressing IFN-α prior to pneumococcal infection demonstrated rapid and effective control of bacterial replication and lung inflammation and improved clinical outcome. Enhanced protection by IFN-α was due to increased activation of neutrophils and macrophages with increased release of reactive oxygen and nitrogen species and bacterial killing. Furthermore, we found that raised levels of IFN-α in the lung remained immune protective even when the gene transfer vector was given at a time postpneumococcal infection. Our study thus shows that the classically antiviral type 1 IFN can be exploited for enhancing immunity against pneumococcal infection via its activating effects on innate immune cells. Our findings hold implications for the therapeutic use of IFN-α gene transfer strategies to combat pneumococcal infections.
Collapse
Affiliation(s)
- Daniela Damjanovic
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University , Hamilton, Ontario, Canada ; M.G. DeGroote Institute for Infectious Disease Research, McMaster University , Hamilton, Ontario, Canada
| | - Amandeep Khera
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University , Hamilton, Ontario, Canada ; M.G. DeGroote Institute for Infectious Disease Research, McMaster University , Hamilton, Ontario, Canada
| | - Maria Fe Medina
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University , Hamilton, Ontario, Canada ; M.G. DeGroote Institute for Infectious Disease Research, McMaster University , Hamilton, Ontario, Canada
| | | | | | - Jack Gauldie
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University , Hamilton, Ontario, Canada ; M.G. DeGroote Institute for Infectious Disease Research, McMaster University , Hamilton, Ontario, Canada
| | - Zhou Xing
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University , Hamilton, Ontario, Canada ; M.G. DeGroote Institute for Infectious Disease Research, McMaster University , Hamilton, Ontario, Canada
| |
Collapse
|
42
|
Jonczyk MS, Simon M, Kumar S, Fernandes VE, Sylvius N, Mallon AM, Denny P, Andrew PW. Genetic factors regulating lung vasculature and immune cell functions associate with resistance to pneumococcal infection. PLoS One 2014; 9:e89831. [PMID: 24594938 PMCID: PMC3940657 DOI: 10.1371/journal.pone.0089831] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Accepted: 01/27/2014] [Indexed: 02/06/2023] Open
Abstract
Streptococcus pneumoniae is an important human pathogen responsible for high mortality and morbidity worldwide. The susceptibility to pneumococcal infections is controlled by as yet unknown genetic factors. To elucidate these factors could help to develop new medical treatments and tools to identify those most at risk. In recent years genome wide association studies (GWAS) in mice and humans have proved successful in identification of causal genes involved in many complex diseases for example diabetes, systemic lupus or cholesterol metabolism. In this study a GWAS approach was used to map genetic loci associated with susceptibility to pneumococcal infection in 26 inbred mouse strains. As a result four candidate QTLs were identified on chromosomes 7, 13, 18 and 19. Interestingly, the QTL on chromosome 7 was located within S. pneumoniae resistance QTL (Spir1) identified previously in a linkage study of BALB/cOlaHsd and CBA/CaOlaHsd F2 intercrosses. We showed that only a limited number of genes encoded within the QTLs carried phenotype-associated polymorphisms (22 genes out of several hundred located within the QTLs). These candidate genes are known to regulate TGFβ signalling, smooth muscle and immune cells functions. Interestingly, our pulmonary histopathology and gene expression data demonstrated, lung vasculature plays an important role in resistance to pneumococcal infection. Therefore we concluded that the cumulative effect of these candidate genes on vasculature and immune cells functions as contributory factors in the observed differences in susceptibility to pneumococcal infection. We also propose that TGFβ-mediated regulation of fibroblast differentiation plays an important role in development of invasive pneumococcal disease. Gene expression data submitted to the NCBI Gene Expression Omnibus Accession No: GSE49533 SNP data submitted to NCBI dbSNP Short Genetic Variation http://www.ncbi.nlm.nih.gov/projects/SNP/snp_viewTable.cgi?handle=MUSPNEUMONIA.
Collapse
Affiliation(s)
- Magda S. Jonczyk
- Department of Infection Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Michelle Simon
- MRC Harwell, Mammalian Genetics Unit, Oxford, United Kingdom
| | - Saumya Kumar
- MRC Harwell, Mammalian Genetics Unit, Oxford, United Kingdom
| | - Vitor E. Fernandes
- Department of Infection Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Nicolas Sylvius
- Department of Genetics, University of Leicester, Leicester, United Kingdom
| | | | - Paul Denny
- MRC Harwell, Mammalian Genetics Unit, Oxford, United Kingdom
| | - Peter W. Andrew
- Department of Infection Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
- * E-mail:
| |
Collapse
|
43
|
Abstract
A 7-valent pneumococcal conjugate vaccine has demonstrated an impact on pneumococcal bacteremia in the immunized pediatric population, extending to nonimmunized adults via herd immunity. A considerable reduction of all-cause pediatric pneumonia has also been found. The impact on all-cause pediatric otitis is limited, but postlicensure data suggests stronger reductions. Higher valency conjugate vaccines are now under development (11V or more). Licensed 23-valent pneumococcal polysaccharide vaccines have been available since 1983 with a demonstrated impact on adult pneumococcal bacteremia. The burden of adult nonbacteremic pneumococcal pneumonia has remained high and efforts to develop improved adult pneumococcal vaccines have been initiated that include conjugates and pneumococcal proteins.
Collapse
Affiliation(s)
- Jan T Poolman
- R & D Bacterial Vaccine Programme, GlaxoSmithKline Biologicals, Rue de l'Institut, 89 1330 Rixensart, Belgium.
| |
Collapse
|
44
|
McKenzie CW, Klonoski JM, Maier T, Trujillo G, Vitiello PF, Huber VC, Lee L. Enhanced response to pulmonary Streptococcus pneumoniae infection is associated with primary ciliary dyskinesia in mice lacking Pcdp1 and Spef2. Cilia 2013; 2:18. [PMID: 24360193 PMCID: PMC3878133 DOI: 10.1186/2046-2530-2-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 12/09/2013] [Indexed: 12/18/2022] Open
Abstract
Background Lower airway abnormalities are common in patients with primary ciliary dyskinesia (PCD), a pediatric syndrome that results from structural or functional defects in motile cilia. Patients can suffer from recurrent bacterial infection in the lung, bronchiectasis, and respiratory distress in addition to chronic sinusitis, otitis media, infertility, and laterality defects. However, surprisingly little is known about the pulmonary phenotype of mouse models of this disorder. Results The pulmonary phenotype of two mouse models of PCD, nm1054 and bgh, which lack Pcdp1 and Spef2, respectively, was investigated by histological and immunohistochemical analysis. In addition, both models were challenged with Streptococcus pneumoniae, a common respiratory pathogen found in the lungs of PCD patients. Histopathological analyses reveal no detectable cellular, developmental, or inflammatory abnormalities in the lower airway of either PCD model. However, exposure to S. pneumoniae results in a markedly enhanced inflammatory response in both models. Based on analysis of inflammatory cells in bronchoalveolar lavage fluid and flow cytometric analysis of cytokines in the lung, the bgh model shows a particularly dramatic lymphocytic response by 3 days post-infection compared to the nm1054 model or wild type animals. Conclusions Defects in ciliary motility result in a severe response to pulmonary infection. The PCD models nm1054 and bgh are distinct and clinically relevant models for future studies investigating the role of mucociliary clearance in host defense.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lance Lee
- Sanford Children's Health Research Center, Sanford Research/USD, 2301 E, 60th St, N, Sioux Falls, SD 57104, USA.
| |
Collapse
|
45
|
Ali YM, Kenawy HI, Muhammad A, Sim RB, Andrew PW, Schwaeble WJ. Human L-ficolin, a recognition molecule of the lectin activation pathway of complement, activates complement by binding to pneumolysin, the major toxin of Streptococcus pneumoniae. PLoS One 2013; 8:e82583. [PMID: 24349316 PMCID: PMC3861440 DOI: 10.1371/journal.pone.0082583] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 11/04/2013] [Indexed: 01/04/2023] Open
Abstract
The complement system is an essential component of the immune response, providing a critical line of defense against different pathogens including S. pneumoniae. Complement is activated via three distinct pathways: the classical (CP), the alternative (AP) and the lectin pathway (LP). The role of Pneumolysin (PLY), a bacterial toxin released by S. pneumoniae, in triggering complement activation has been studied in vitro. Our results demonstrate that in both human and mouse sera complement was activated via the CP, initiated by direct binding of even non-specific IgM and IgG3 to PLY. Absence of CP activity in C1q(-/-) mouse serum completely abolished any C3 deposition. However, C1q depleted human serum strongly opsonized PLY through abundant deposition of C3 activation products, indicating that the LP may have a vital role in activating the human complement system on PLY. We identified that human L-ficolin is the critical LP recognition molecule that drives LP activation on PLY, while all of the murine LP recognition components fail to bind and activate complement on PLY. This work elucidates the detailed interactions between PLY and complement and shows for the first time a specific role of the LP in PLY-mediated complement activation in human serum.
Collapse
Affiliation(s)
- Youssif M. Ali
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
- Department of Microbiology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Hany I. Kenawy
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
- Department of Microbiology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Adnan Muhammad
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Robert B. Sim
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Peter W. Andrew
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Wilhelm J. Schwaeble
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
46
|
Herrera M, Salva S, Villena J, Barbieri N, Alvarez S. Lactobacillus rhamnosus CRL1505 enhances systemic and respiratory innate immune response in immunocompromised malnourished mice. J Funct Foods 2013. [DOI: 10.1016/j.jff.2013.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
47
|
Marked Improvement of Severe Lung Immunopathology by Influenza-Associated Pneumococcal Superinfection Requires the Control of Both Bacterial Replication and Host Immune Responses. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:868-80. [DOI: 10.1016/j.ajpath.2013.05.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Revised: 04/21/2013] [Accepted: 05/13/2013] [Indexed: 12/19/2022]
|
48
|
Otero C, Paz RD, Galassi N, Bezrodnik L, Finiasz MR, Fink S. Immune response to Streptococcus pneumoniae in asthma patients: comparison between stable situation and exacerbation. Clin Exp Immunol 2013; 173:92-101. [PMID: 23607482 DOI: 10.1111/cei.12082] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2013] [Indexed: 01/01/2023] Open
Abstract
In Argentina, more than 3 million people suffer from asthma, with numbers rising. When asthma patients acquire viral infections which, in turn, trigger the asthmatic response, they may develop subsequent bacterial infections, mainly by Streptococcus (S.) pneumoniae. This encapsulated Gram(+) bacterium has been considered historically a T cell-independent antigen. Nevertheless, several papers describe the role of T cells in the immune response to S. pneumoniae. We evaluated the response to S. pneumoniae and compared it to the response to Mycobacterium (M.) tuberculosis, a different type of bacterium that requires a T helper type 1 (Th1) response, in cells from atopic asthmatic children, to compare parameters for the same individual under exacerbation and in a stable situation whenever possible. We studied asthma patients and a control group of age-matched children, evaluating cell populations, activation markers and cytokine production by flow cytometry, and cytokine concentration in serum and cell culture supernatants by enzyme-linked immunosorbent assay (ELISA). No differences were observed in γδ T cells for the same patient in either situation, and a tendency to lower percentages of CD4(+) CD25(hi) T cells was observed under stability. A significantly lower production of tumour necrosis factor (TNF)-α and a significantly higher production of interleukin (IL)-5 was observed in asthma patients compared to healthy individuals, but no differences could be observed for IL-4, IL-13 or IL-10. A greater early activation response against M. tuberculosis, compared to S. pneumoniae, was observed in the asthmatic patients' cells. This may contribute to explaining why these patients frequently acquire infections caused by the latter bacterium and not the former.
Collapse
Affiliation(s)
- C Otero
- Immune Response to Human Infections Laboratory, IMEX-CONICET-Academia Nacional de Medicina, Argentina
| | | | | | | | | | | |
Collapse
|
49
|
Rosendahl A, Bergmann S, Hammerschmidt S, Goldmann O, Medina E. Lung dendritic cells facilitate extrapulmonary bacterial dissemination during pneumococcal pneumonia. Front Cell Infect Microbiol 2013; 3:21. [PMID: 23802100 PMCID: PMC3689026 DOI: 10.3389/fcimb.2013.00021] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 06/04/2013] [Indexed: 11/16/2022] Open
Abstract
Streptococcus pneumoniae is a leading cause of bacterial pneumonia worldwide. Given the critical role of dendritic cells (DCs) in regulating and modulating the immune response to pathogens, we investigated here the role of DCs in S. pneumoniae lung infections. Using a well-established transgenic mouse line which allows the conditional transient depletion of DCs, we showed that ablation of DCs resulted in enhanced resistance to intranasal challenge with S. pneumoniae. DCs-depleted mice exhibited delayed bacterial systemic dissemination, significantly reduced bacterial loads in the infected organs and lower levels of serum inflammatory mediators than non-depleted animals. The increased resistance of DCs-depleted mice to S. pneumoniae was associated with a better capacity to restrict pneumococci extrapulmonary dissemination. Furthermore, we demonstrated that S. pneumoniae disseminated from the lungs into the regional lymph nodes in a cell-independent manner and that this direct way of dissemination was much more efficient in the presence of DCs. We also provide evidence that S. pneumoniae induces expression and activation of matrix metalloproteinase-9 (MMP-9) in cultured bone marrow-derived DCs. MMP-9 is a protease involved in the breakdown of extracellular matrix proteins and is critical for DC trafficking across extracellular matrix and basement membranes during the migration from the periphery to the lymph nodes. MMP-9 was also significantly up-regulated in the lungs of mice after intranasal infection with S. pneumoniae. Notably, the expression levels of MMP-9 in the infected lungs were significantly decreased after depletion of DCs suggesting the involvement of DCs in MMP-9 production during pneumococcal pneumonia. Thus, we propose that S. pneumoniae can exploit the DC-derived proteolysis to open tissue barriers thereby facilitating its own dissemination from the local site of infection.
Collapse
Affiliation(s)
- Alva Rosendahl
- Infection Immunology Research Group, Department of Medical Microbiology, Helmholtz Centre for Infection ResearchBraunschweig, Germany
| | - Simone Bergmann
- Institute for Microbiology, Department of Cellular Infection Biology, Technische UniversitätBraunschweig, Germany
| | - Sven Hammerschmidt
- Department Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz University of GreifswaldGreifswald, Germany
| | - Oliver Goldmann
- Infection Immunology Research Group, Department of Medical Microbiology, Helmholtz Centre for Infection ResearchBraunschweig, Germany
| | - Eva Medina
- Infection Immunology Research Group, Department of Medical Microbiology, Helmholtz Centre for Infection ResearchBraunschweig, Germany
| |
Collapse
|
50
|
Ivanov S, Renneson J, Fontaine J, Barthelemy A, Paget C, Fernandez EM, Blanc F, De Trez C, Van Maele L, Dumoutier L, Huerre MR, Eberl G, Si-Tahar M, Gosset P, Renauld JC, Sirard JC, Faveeuw C, Trottein F. Interleukin-22 reduces lung inflammation during influenza A virus infection and protects against secondary bacterial infection. J Virol 2013; 87:6911-24. [PMID: 23596287 PMCID: PMC3676141 DOI: 10.1128/jvi.02943-12] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 04/04/2013] [Indexed: 12/30/2022] Open
Abstract
Interleukin-22 (IL-22) has redundant, protective, or pathogenic functions during autoimmune, inflammatory, and infectious diseases. Here, we addressed the potential role of IL-22 in host defense and pathogenesis during lethal and sublethal respiratory H3N2 influenza A virus (IAV) infection. We show that IL-22, as well as factors associated with its production, are expressed in the lung tissue during the early phases of IAV infection. Our data indicate that retinoic acid receptor-related orphan receptor-γt (RORγt)-positive αβ and γδ T cells, as well as innate lymphoid cells, expressed enhanced Il22 transcripts as early as 2 days postinfection. During lethal or sublethal IAV infections, endogenous IL-22 played no role in the control of IAV replication and in the development of the IAV-specific CD8(+) T cell response. During lethal infection, where wild-type (WT) mice succumbed to severe pneumonia, the lack of IL-22 did not accelerate or delay IAV-associated pathogenesis and animal death. In stark contrast, during sublethal IAV infection, IL-22-deficient animals had enhanced lung injuries and showed a lower airway epithelial integrity relative to WT littermates. Of importance, the protective effect of endogenous IL-22 in pulmonary damages was associated with a more controlled secondary bacterial infection. Indeed, after challenge with Streptococcus pneumoniae, IAV-experienced Il22(-/-) animals were more susceptible than WT controls in terms of survival rate and bacterial burden in the lungs. Together, IL-22 plays no major role during lethal influenza but is beneficial during sublethal H3N2 IAV infection, where it limits lung inflammation and subsequent bacterial superinfections.
Collapse
Affiliation(s)
- Stoyan Ivanov
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Université Lille Nord de France, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - Joelle Renneson
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Université Lille Nord de France, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - Josette Fontaine
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Université Lille Nord de France, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - Adeline Barthelemy
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Université Lille Nord de France, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - Christophe Paget
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Université Lille Nord de France, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - Elodie Macho Fernandez
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Université Lille Nord de France, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - Fany Blanc
- Institut Pasteur, Paris, France
- Institut National de la Santé et de la Recherche Médicale, U874, Paris, France
| | - Carl De Trez
- Vlaams Interuniversitair Instituut voor Biotechnologie, Vrije Universiteit Brussel, Brussels, Belgium
| | - Laurye Van Maele
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Université Lille Nord de France, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - Laure Dumoutier
- Ludwig Institute for Cancer Research, Université Catholique de Louvain, Brussels
| | - Michel-René Huerre
- Institut Pasteur, Paris, France
- Unite de Recherche et d'Expertise Histotechnologie et Pathologie, Paris, France
| | - Gérard Eberl
- Institut Pasteur, Paris, France
- Centre National de la Recherche Scientifique, URA 1961, Paris, France
| | - Mustapha Si-Tahar
- Institut Pasteur, Paris, France
- Institut National de la Santé et de la Recherche Médicale, U874, Paris, France
| | - Pierre Gosset
- Hopital Saint Vincent, Groupe Hospitalier de l'Institut Catholique de Lille, Université Catholique de Lille, Lille, France
| | | | - Jean Claude Sirard
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Université Lille Nord de France, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - Christelle Faveeuw
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Université Lille Nord de France, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - François Trottein
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Université Lille Nord de France, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| |
Collapse
|