51
|
Differential IL-17A response to S. pneumoniae in adenoid tissue of children with sleep disordered breathing and otitis media with effusion. Sci Rep 2019; 9:19839. [PMID: 31882693 PMCID: PMC6934741 DOI: 10.1038/s41598-019-56415-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 11/18/2019] [Indexed: 12/25/2022] Open
Abstract
Streptococcus pneumonia, one of the major colonizers in nasopharyngeal adenoids, has been the predominant pathogen causing acute otitis media (AOM) in children. Recent evidence suggests an association between IL-17A-mediated immune response and the clearance of pneumococcal colonization in nasopharyngeal adenoids. Here, we evaluated the expressions of IL-17A and associated genes in hypertrophic adenoid tissues of children with sleep-disordered breathing (SDB) and otitis media with effusion (OME) and their association with pneumococcal carriage. Sixty-six pediatric patients with adenoid hypertrophy were enrolled. During adenoidectomy, nasopharyngeal swab and adenoid tissues were used to determine pneumococcal carriage and IL-17A expression. Our results revealed significantly higher levels of IL-17A and IL-17A:IL-10 mRNA in the SDB patients positive for nasopharyngeal pneumococcal carriage than those negative. However, these differences were not significant in the OME group. These results suggested, in OME patients, prolonged or chronic pneumococcal carriage may occur because of insufficient IL-17A-mediated mucosal clearance, and could further lead to AOM and OME development.
Collapse
|
52
|
Combined prime-boost immunization with systemic and mucosal pneumococcal vaccines based on Pneumococcal surface protein A to enhance protection against lethal pneumococcal infections. Immunol Res 2019; 67:398-407. [PMID: 31773490 DOI: 10.1007/s12026-019-09107-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Limited protective effects of commercially available vaccines necessitate the development of novel pneumococcal vaccines. We recently reported a pneumococcal systemic vaccine containing two proteins, Pneumococcal surface protein A (PspA of family 1 and 2) and a bacterium-like particle-based pneumococcal mucosal vaccine containing PspA2 and PspA4 fragments, both eliciting broad protective immune responses. We had previously reported that subcutaneous (s.c.+s.c.+s.c.) immunization with the systemic vaccine induced more pronounced humoral serum IgG responses, while intranasal (i.n.+i.n.+i.n.) immunization with the mucosal vaccine elicited a more pronounced mucosal secretory IgA (sIgA) response. We hypothesized that a combinatorial administration of the two vaccines might elicit more pronounced and broader protective immune responses. Therefore, this study aimed to determine the efficacy of combinatorial prime-boost immunization using both systemic and mucosal vaccines for a pneumococcal infection. Combinatorial prime-boost immunization (s.c.+i.n. and i.n.+s.c.) induced not only IgG, but also mucosal sIgA production at high levels. Systemic priming and mucosal boosting immunization (s.c.+i.n.) provided markedly better protection than homologous prime-boost immunization (s.c.+s.c.+s.c. and i.n.+i.n.+i.n.). Moreover, it induced more robust Th1 and Th17 cell-mediated immune responses than mucosal priming and systemic boosting immunization (i.n.+s.c.). These results indicate that combinatorial prime-boost immunization potentially induces a robust systemic and mucosal immune response, making it an optimal alternative for maximum protection against lethal pneumococcal infections.
Collapse
|
53
|
Opening the OPK Assay Gatekeeper: Harnessing Multi-Modal Protection by Pneumococcal Vaccines. Pathogens 2019; 8:pathogens8040203. [PMID: 31652741 PMCID: PMC6963391 DOI: 10.3390/pathogens8040203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 10/14/2019] [Accepted: 10/21/2019] [Indexed: 11/16/2022] Open
Abstract
Pneumococcal vaccine development is driven by the achievement of high activity in a single gatekeeper assay: the bacterial opsonophagocytic killing (OPK) assay. New evidence challenges the dogma that anti-capsular antibodies have only a single function that predicts success. The emerging concept of multi-modal protection presents an array of questions that are fundamental to adopting a new vaccine design process. If antibodies have hidden non-opsonic functions that are protective, should these be optimized for better vaccines? What would protein antigens add to protective activity? Are cellular immune functions additive to antibodies for success? Do different organs benefit from different modes of protection? Can vaccine activities beyond OPK protect the immunocompromised host? This commentary raises these issues at a time when capsule-only OPK assay-based vaccines are increasingly seen as a limiting strategy.
Collapse
|
54
|
Collini PJ, Bewley MA, Mohasin M, Marriott HM, Miller RF, Geretti AM, Beloukas A, Papadimitropoulos A, Read RC, Noursadeghi M, Dockrell DH. HIV gp120 in the Lungs of Antiretroviral Therapy-treated Individuals Impairs Alveolar Macrophage Responses to Pneumococci. Am J Respir Crit Care Med 2019; 197:1604-1615. [PMID: 29365279 DOI: 10.1164/rccm.201708-1755oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
RATIONALE People living with HIV are at significantly increased risk of invasive pneumococcal disease, despite long-term antiretroviral therapy (ART). The mechanism explaining this observation remains undefined. OBJECTIVES To determine if apoptosis-associated microbicidal mechanisms, required to clear intracellular pneumococci that survive initial phagolysosomal killing, are perturbed. METHODS Alveolar macrophages (AM) were obtained by BAL from healthy donors or HIV-1-seropositive donors on long-term ART with undetectable plasma viral load. Monocyte-derived macrophages (MDM) were obtained from healthy donors and infected with HIV-1BaL or treated with gp120. Macrophages were challenged with opsonized serotype 2 Streptococcus pneumoniae and assessed for apoptosis, bactericidal activity, protein expression, and mitochondrial reactive oxygen species (mROS). AM phenotyping, ultrasensitive HIV-1 RNA quantification, and gp120 measurement were also performed in BAL. MEASUREMENTS AND MAIN RESULTS HIV-1BaL infection impaired apoptosis, induction of mROS, and pneumococcal killing by MDM. Apoptosis-associated pneumococcal killing was also reduced in AM from ART-treated HIV-1-seropositive donors. BAL fluid from these individuals demonstrated persistent lung CD8+ T lymphocytosis, and gp120 or HIV-1 RNA was also detected. Despite this, transcriptional activity in AM freshly isolated from people living with HIV was broadly similar to healthy volunteers. Instead, gp120 phenocopied the defect in pneumococcal killing in healthy MDM through post-translational modification of Mcl-1, preventing apoptosis induction, caspase activation, and increased mROS generation. Moreover, gp120 also inhibited mROS-dependent pneumococcal killing in MDM. CONCLUSIONS Despite ART, HIV-1, via gp120, drives persisting innate immune defects in AM microbicidal mechanisms, enhancing susceptibility to pneumococcal disease.
Collapse
Affiliation(s)
- Paul J Collini
- 1 The Florey Institute for Host-Pathogen Interactions and Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom.,2 Academic Directorate of Communicable Diseases and Specialised Medicine, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Martin A Bewley
- 1 The Florey Institute for Host-Pathogen Interactions and Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Mohamed Mohasin
- 1 The Florey Institute for Host-Pathogen Interactions and Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Helen M Marriott
- 1 The Florey Institute for Host-Pathogen Interactions and Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom
| | - Robert F Miller
- 3 Research Department of Infection and Population Health, Institute of Epidemiology & Health Care, Faculty of Population Health Sciences, and
| | - Anna-Maria Geretti
- 4 Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Apostolos Beloukas
- 4 Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Athanasios Papadimitropoulos
- 4 Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Robert C Read
- 5 Academic Unit of Clinical and Experimental Sciences, University of Southampton and National Institute for Health Research Southampton Biomedical Research Centre, Southampton, United Kingdom; and
| | - Mahdad Noursadeghi
- 6 Division of Infection & Immunity, Faculty of Medical Sciences, University College London, London, United Kingdom
| | - David H Dockrell
- 1 The Florey Institute for Host-Pathogen Interactions and Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield Medical School, Sheffield, United Kingdom.,2 Academic Directorate of Communicable Diseases and Specialised Medicine, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom.,7 MRC/UoE Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
55
|
Jochems SP, de Ruiter K, Solórzano C, Voskamp A, Mitsi E, Nikolaou E, Carniel BF, Pojar S, German EL, Reiné J, Soares-Schanoski A, Hill H, Robinson R, Hyder-Wright AD, Weight CM, Durrenberger PF, Heyderman RS, Gordon SB, Smits HH, Urban BC, Rylance J, Collins AM, Wilkie MD, Lazarova L, Leong SC, Yazdanbakhsh M, Ferreira DM. Innate and adaptive nasal mucosal immune responses following experimental human pneumococcal colonization. J Clin Invest 2019; 129:4523-4538. [PMID: 31361601 PMCID: PMC6763269 DOI: 10.1172/jci128865] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Streptococcus pneumoniae (Spn) is a common cause of respiratory infection, but also frequently colonizes the nasopharynx in the absence of disease. We used mass cytometry to study immune cells from nasal biopsy samples collected following experimental human pneumococcal challenge in order to identify immunological mechanisms of control of Spn colonization. Using 37 markers, we characterized 293 nasal immune cell clusters, of which 7 were associated with Spn colonization. B cell and CD161+CD8+ T cell clusters were significantly lower in colonized than in noncolonized subjects. By following a second cohort before and after pneumococcal challenge we observed that B cells were depleted from the nasal mucosa upon Spn colonization. This associated with an expansion of Spn polysaccharide–specific and total plasmablasts in blood. Moreover, increased responses of blood mucosa-associated invariant T (MAIT) cells against in vitro stimulation with pneumococcus prior to challenge associated with protection against establishment of Spn colonization and with increased mucosal MAIT cell populations. These results implicate MAIT cells in the protection against pneumococcal colonization and demonstrate that colonization affects mucosal and circulating B cell populations.
Collapse
Affiliation(s)
- Simon P Jochems
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom.,Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Karin de Ruiter
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Carla Solórzano
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Astrid Voskamp
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Elena Mitsi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Elissavet Nikolaou
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Beatriz F Carniel
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Sherin Pojar
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Esther L German
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Jesús Reiné
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | | | - Helen Hill
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom.,Royal Liverpool and Broadgreen University Hospital, Liverpool, United Kingdom
| | - Rachel Robinson
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom.,Royal Liverpool and Broadgreen University Hospital, Liverpool, United Kingdom
| | - Angela D Hyder-Wright
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom.,Royal Liverpool and Broadgreen University Hospital, Liverpool, United Kingdom
| | | | - Pascal F Durrenberger
- Centre for Inflammation and Tissue Repair, University College London, London, United Kingdom
| | | | - Stephen B Gordon
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom.,Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - Hermelijn H Smits
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Britta C Urban
- Department of Parasitology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Jamie Rylance
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Andrea M Collins
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom.,Royal Liverpool and Broadgreen University Hospital, Liverpool, United Kingdom.,Aintree University Hospital NHS Foundation Trust, Liverpool, United Kingdom
| | - Mark D Wilkie
- Royal Liverpool and Broadgreen University Hospital, Liverpool, United Kingdom
| | - Lepa Lazarova
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom.,Royal Liverpool and Broadgreen University Hospital, Liverpool, United Kingdom
| | - Samuel C Leong
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom.,Department of Otorhinolaryngology - Head and Neck Surgery, Aintree University Hospital NHS Foundation Trust, Liverpool, United Kingdom
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Daniela M Ferreira
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| |
Collapse
|
56
|
Subramanian K, Henriques-Normark B, Normark S. Emerging concepts in the pathogenesis of the Streptococcus pneumoniae: From nasopharyngeal colonizer to intracellular pathogen. Cell Microbiol 2019; 21:e13077. [PMID: 31251447 PMCID: PMC6899785 DOI: 10.1111/cmi.13077] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 06/17/2019] [Accepted: 06/21/2019] [Indexed: 12/19/2022]
Abstract
Streptococcus pneumoniae (the pneumococcus) is a human respiratory tract pathogen and a major cause of morbidity and mortality globally. Although the pneumococcus is a commensal bacterium that colonizes the nasopharynx, it also causes lethal diseases such as meningitis, sepsis, and pneumonia, especially in immunocompromised patients, in the elderly, and in young children. Due to the acquisition of antibiotic resistance and the emergence of nonvaccine serotypes, the pneumococcus has been classified as one of the priority pathogens for which new antibacterials are urgently required by the World Health Organization, 2017. Understanding molecular mechanisms behind the pathogenesis of pneumococcal infections and bacterial interactions within the host is crucial to developing novel therapeutics. Previously considered to be an extracellular pathogen, it is becoming evident that pneumococci may also occasionally establish intracellular niches within the body to escape immune surveillance and spread within the host. Intracellular survival within host cells also enables pneumococci to resist many antibiotics. Within the host cell, the bacteria exist in unique vacuoles, thereby avoiding degradation by the acidic lysosomes, and modulate the expression of its virulence genes to adapt to the intracellular environment. To invade and survive intracellularly, the pneumococcus utilizes a combination of virulence factors such as pneumolysin (PLY), pneumococcal surface protein A (PspA), pneumococcal adhesion and virulence protein B (PavB), the pilus‐1 adhesin RrgA, pyruvate oxidase (SpxB), and metalloprotease (ZmpB). In this review, we discuss recent findings showing the intracellular persistence of Streptococcus pneumoniae and its underlying mechanisms.
Collapse
Affiliation(s)
- Karthik Subramanian
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Birgitta Henriques-Normark
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden.,Lee Kong Chian School of Medicine (LKC) and Singapore Centre on Environmental Life Sciences Engineering (SCELSE), Nanyang Technological University, Singapore
| | - Staffan Normark
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden.,Lee Kong Chian School of Medicine (LKC) and Singapore Centre on Environmental Life Sciences Engineering (SCELSE), Nanyang Technological University, Singapore
| |
Collapse
|
57
|
Feng S, Chen T, Lei G, Hou F, Jiang J, Huang Q, Peng Y, Ye C, Hu DL, Fang R. Absent in melanoma 2 inflammasome is required for host defence against Streptococcus pneumoniae infection. Innate Immun 2019; 25:412-419. [PMID: 31266383 PMCID: PMC6900643 DOI: 10.1177/1753425919860252] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Streptococcus pneumoniae, a leading cause of invasive
pneumococcal disease, is responsible for high mortality and morbidity worldwide.
A previous study showed that the NLR family pyrin domain containing 3 (NLRP3)
and absent in melanoma 2 (AIM2) inflammasomes are essential for caspase-1
activation and IL-1β production in the host response to S.
pneumoniae infection. The function of NLRP3 in host innate immunity
to S. pneumoniae was studied in vivo and
in vitro. However, the role of AIM2 in host defence against
S. pneumoniae remains unclear. Here, we show that
AIM2-deficient (AIM2–/–) mice display increased susceptibility to
intra-nasal infection with S. pneumoniae in comparison to wild
type mice and that this susceptibility was associated with defective IL-1β
production. Macrophages from AIM2–/– mice infected with S.
pneumoniae showed impaired secretion of IL-1β as well as activation
of the inflammasome, as determined by the oligomerisation of
apoptosis-associated speck-like protein containing a CARD (ASC) and caspase-1
activation. Taken together, these results indicate that the AIM2 inflammasome is
essential for caspase-1-dependent cytokine IL-1β production and eventual
protection from pneumococcal infection in mice.
Collapse
Affiliation(s)
- Siwei Feng
- 1 College of Animal Science and Technology, Southwest University, PR China
| | - Tingting Chen
- 1 College of Animal Science and Technology, Southwest University, PR China
| | - Guihua Lei
- 1 College of Animal Science and Technology, Southwest University, PR China
| | - Fengqing Hou
- 1 College of Animal Science and Technology, Southwest University, PR China
| | - Jiali Jiang
- 1 College of Animal Science and Technology, Southwest University, PR China
| | - Qingyuan Huang
- 1 College of Animal Science and Technology, Southwest University, PR China
| | - Yuanyi Peng
- 1 College of Animal Science and Technology, Southwest University, PR China
| | - Chao Ye
- 1 College of Animal Science and Technology, Southwest University, PR China
| | - Dong-Liang Hu
- 1 College of Animal Science and Technology, Southwest University, PR China.,2 Department of Zoonoses, Kitasato University School of Veterinary Medicine, Japan
| | - Rendong Fang
- 1 College of Animal Science and Technology, Southwest University, PR China
| |
Collapse
|
58
|
Prediction and Validation of Immunogenic Domains of Pneumococcal Proteins Recognized by Human CD4 + T Cells. Infect Immun 2019; 87:IAI.00098-19. [PMID: 30910792 PMCID: PMC6529658 DOI: 10.1128/iai.00098-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 02/28/2019] [Indexed: 12/24/2022] Open
Abstract
CD4+ T-cell mechanisms are implied in protection against pneumococcal colonization; however, their target antigens and function are not well defined. In contrast to high-throughput protein arrays for serology, basic antigen tools for CD4+ T-cell studies are lacking. CD4+ T-cell mechanisms are implied in protection against pneumococcal colonization; however, their target antigens and function are not well defined. In contrast to high-throughput protein arrays for serology, basic antigen tools for CD4+ T-cell studies are lacking. Here, we evaluate the potential of a bioinformatics tool for in silico prediction of immunogenicity as a method to reveal domains of pneumococcal proteins targeted by human CD4+ T cells. For 100 pneumococcal proteins, CD4+ T-cell immunogenicity was predicted based on HLA-DRB1 binding motifs. For 20 potentially CD4+ T-cell immunogenic proteins, epitope regions were verified by testing synthetic peptides in T-cell assays using peripheral blood mononuclear cells from healthy adults. Peptide pools of 19 out of 20 proteins evoked T-cell responses. The most frequent responses (detectable in ≥20% of donors tested) were found to SP_0117 (PspA), SP_0468 (putative sortase), SP_0546 (BlpZ), SP_1650 (PsaA), SP_1923 (Ply), SP_2048 (conserved hypothetical protein), SP_2216 (PscB), and SPR_0907 (PhtD). Responding donors had diverging recognition patterns and profiles of signature cytokines (gamma interferon [IFN-γ], tumor necrosis factor alpha [TNF-α], interleukin-13 [IL-13], and/or IL-17A) against single-epitope regions. Natural HLA-DR-restricted presentation and recognition of a predicted SP_1923-derived epitope were validated through the isolation of a CD4+ T-cell clone producing IFN-γ, TNF-α, and IL-17A in response to the synthetic peptide, whole protein, and heat-inactivated pneumococcus. This proof of principle for a bioinformatics tool to identify pneumococcal protein epitopes targeted by human CD4+ T cells provides a peptide-based strategy to study cell-mediated immune mechanisms for the pneumococcal proteome, advancing the development of immunomonitoring assays and targeted vaccine approaches.
Collapse
|
59
|
Riegler AN, Brissac T, Gonzalez-Juarbe N, Orihuela CJ. Necroptotic Cell Death Promotes Adaptive Immunity Against Colonizing Pneumococci. Front Immunol 2019; 10:615. [PMID: 31019504 PMCID: PMC6459137 DOI: 10.3389/fimmu.2019.00615] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/08/2019] [Indexed: 12/12/2022] Open
Abstract
Pore-forming toxin (PFT) induced necroptosis exacerbates pulmonary injury during bacterial pneumonia. However, its role during asymptomatic nasopharyngeal colonization and toward the development of protective immunity was unknown. Using a mouse model of Streptococcus pneumoniae (Spn) asymptomatic colonization, we determined that nasopharyngeal epithelial cells (nEC) died of pneumolysin (Ply)-dependent necroptosis. Mice deficient in MLKL, the necroptosis effector, or challenged with Ply-deficient Spn showed less nEC sloughing, increased neutrophil infiltration, and altered IL-1α, IL-33, CXCL2, IL-17, and IL-6 levels in nasal lavage fluid (NALF). Activated MLKL correlated with increased presence of CD11c+ antigen presenting cells in Spn-associated submucosa. Colonized MLKL KO mice and wildtype mice colonized with Ply-deficient Spn produced less antibody against the bacterial surface protein PspA, were delayed in bacterial clearance, and were more susceptible to a lethal secondary Spn challenge. We conclude that PFT-induced necroptosis is instrumental in the natural development of protective immunity against opportunistic PFT-producing bacterial pathogens.
Collapse
Affiliation(s)
- Ashleigh Nichole Riegler
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Terry Brissac
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Norberto Gonzalez-Juarbe
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Carlos J Orihuela
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
60
|
Johnson LR, Vernau W. Bronchoalveolar lavage fluid lymphocytosis in 104 dogs (2006-2016). J Vet Intern Med 2019; 33:1315-1321. [PMID: 30912207 PMCID: PMC6524393 DOI: 10.1111/jvim.15489] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/15/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Bronchoalveolar lavage (BAL) fluid cytology and culture are used to characterize respiratory diseases in dogs. Little is known about disorders associated with increased numbers of lymphocytes in BAL fluid. OBJECTIVE To evaluate duration of clinical signs and detection of specific respiratory diagnoses in dogs with BAL lymphocytosis. ANIMALS One-hundred four client-owned dogs evaluated for respiratory signs. METHODS Medical records of dogs that had >300 cells/μL and >20% lymphocytes on a differential cell count of BAL fluid between January 1, 2006, and January 1, 2016, were reviewed retrospectively. Cases were evaluated for the duration of clinical signs and respiratory diagnoses, including aspiration injury, infectious or inflammatory respiratory disease, and airway collapse. RESULTS Dogs ranged in age from 0.5 to 16 years (median, 7.9 years) and had a median body weight of 11.4 kg (range, 2.0-42.7 kg). Eosinophilic lung disease was documented in 13 of 104 dogs (Group 1) and airway neutrophilia associated with infectious or inflammatory disease was found in 59 of 104 dogs (Group 2). Lymphocytosis alone in BAL fluid was described in 32 dogs (Group 3). Duration of cough did not differ among groups, but airway collapse was significantly more common in dogs with solitary lymphocytosis than in those with other types of inflammation. CONCLUSIONS AND CLINICAL IMPORTANCE Lymphocytosis in BAL fluid is common in dogs and, in many cases, likely represents a common response to airway injury, independent of the type or duration of insult. It is unknown whether airway collapse leads to lymphocytosis or if the inflammatory process causes airway collapse.
Collapse
Affiliation(s)
- Lynelle R Johnson
- Department of Medicine and Epidemiology, University of California School of Veterinary Medicine, Davis, California
| | - William Vernau
- Department of Pathology, Microbiology and Immunology, University of California School of Veterinary Medicine, Davis, California
| |
Collapse
|
61
|
Intranasal Immunization with the Commensal Streptococcus mitis Confers Protective Immunity against Pneumococcal Lung Infection. Appl Environ Microbiol 2019; 85:AEM.02235-18. [PMID: 30683742 DOI: 10.1128/aem.02235-18] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/02/2019] [Indexed: 02/04/2023] Open
Abstract
Streptococcus pneumoniae is a bacterial pathogen that causes various diseases of public health concern worldwide. Current pneumococcal vaccines target the capsular polysaccharide surrounding the cells. However, only up to 13 of more than 90 pneumococcal capsular serotypes are represented in the current conjugate vaccines. In this study, we used two experimental approaches to evaluate the potential of Streptococcus mitis, a commensal that exhibits immune cross-reactivity with S. pneumoniae, to confer protective immunity to S. pneumoniae lung infection in mice. First, we assessed the immune response and protective effect of wild-type S. mitis against lung infection by S. pneumoniae strains D39 (serotype 2) and TIGR4 (serotype 4). Second, we examined the ability of an S. mitis mutant expressing the S. pneumoniae type 4 capsule (S. mitis TIGR4cps) to elicit focused protection against S. pneumoniae TIGR4. Our results showed that intranasal immunization of mice with S. mitis produced significantly higher levels of serum IgG and IgA antibodies reactive to both S. mitis and S. pneumoniae, as well as enhanced production of interleukin 17A (IL-17A), but not gamma interferon (IFN-γ) and IL-4, compared with control mice. The immunization resulted in a reduced bacterial load in respiratory tissues following lung infection with S. pneumoniae TIGR4 or D39 compared with control mice. With S. mitis TIGR4cps, protection upon challenge with S. pneumoniae TIGR4 was superior. Thus, these findings show the potential of S. mitis to elicit natural serotype-independent protection against two pneumococcal serotypes and to provide the benefits of the well-recognized protective effect of capsule-targeting vaccines.IMPORTANCE Streptococcus pneumoniae causes various diseases worldwide. Current pneumococcal vaccines protect against a limited number of more than 90 pneumococcal serotypes, accentuating the urgent need to develop novel prophylactic strategies. S. pneumoniae and the commensal Streptococcus mitis share immunogenic characteristics that make S. mitis an attractive vaccine candidate against S. pneumoniae In this study, we evaluated the potential of S. mitis and its mutant expressing pneumococcal capsule type 4 (S. mitis TIGR4cps) to induce protection against S. pneumoniae lung infection in mice. Our findings show that intranasal vaccination with S. mitis protects against S. pneumoniae strains D39 (serotype 2) and TIGR4 (serotype 4) in a serotype-independent fashion, which is associated with enhanced antibody and T cell responses. Furthermore, S. mitis TIGR4cps conferred additional protection against S. pneumoniae TIGR4, but not against D39. The findings highlight the potential of S. mitis to generate protection that combines both serotype-independent and serotype-specific responses.
Collapse
|
62
|
Ramos-Sevillano E, Ercoli G, Brown JS. Mechanisms of Naturally Acquired Immunity to Streptococcus pneumoniae. Front Immunol 2019; 10:358. [PMID: 30881363 PMCID: PMC6405633 DOI: 10.3389/fimmu.2019.00358] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 02/12/2019] [Indexed: 12/11/2022] Open
Abstract
In this review we give an update on the mechanisms of naturally acquired immunity against Streptococcus pneumoniae, one of the major human bacterial pathogens that is a common cause of pneumonia, septicaemia, and meningitis. A clear understanding of the natural mechanisms of immunity to S. pneumoniae is necessary to help define why the very young and elderly are at high risk of disease, and for devising new prevention strategies. Recent data has shown that nasopharynx colonization by S. pneumoniae induces antibody responses to protein and capsular antigens in both mice and humans, and also induces Th17 CD4+ cellular immune responses in mice and increases pre-existing responses in humans. These responses are protective, demonstrating that colonization is an immunizing event. We discuss the data from animal models and humans on the relative importance of naturally acquired antibody and Th17 cells on immunity to S. pneumoniae at three different anatomical sites of infection, the nasopharynx (the site of natural asymptomatic carriage), the lung (site of pneumonia), and the blood (site of sepsis). Mouse data suggest that CD4+ Th17 cells prevent both primary and secondary nasopharyngeal carriage with no role for antibody induced by previous colonization. In contrast, antibody is necessary for prevention of sepsis but CD4+ cellular responses are not. Protection against pneumonia requires a combination of both antibody and Th17 cells, in both cases targeting protein rather than capsular antigen. Proof of which immune component prevents human infection is less easily available, but two recent papers demonstrate that human IgG targeting S. pneumoniae protein antigens is highly protective against septicaemia. The role of CD4+ responses to prior nasopharyngeal colonization for protective immunity in humans is unclear. The evidence that there is significant naturally-acquired immunity to S. pneumoniae independent of anti-capsular polysaccharide has clinical implications for the detection of subjects at risk of S. pneumoniae infections, and the data showing the importance of protein antigens as targets for antibody and Th17 mediated immunity should aid the development of new vaccine strategies.
Collapse
Affiliation(s)
| | | | - Jeremy S. Brown
- Centre for Inflammation and Tissue Repair, UCL Respiratory, London, United Kingdom
| |
Collapse
|
63
|
Neonatal intestinal immune regulation by the commensal bacterium, P. UF1. Mucosal Immunol 2019; 12:434-444. [PMID: 30647410 PMCID: PMC6375783 DOI: 10.1038/s41385-018-0125-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/23/2018] [Accepted: 12/07/2018] [Indexed: 02/04/2023]
Abstract
Newborns are highly susceptible to pathogenic infections with significant worldwide morbidity possibly due to an immature immune system. Recently, we reported that Propionibacterium strain, P. UF1, isolated from the gut microbiota of preterm infants, induced the differentiation of bacteria-specific Th17 cells. Here, we demonstrate that P. UF1 significantly increased the number of protective Th17 cells and maintained IL-10+ regulatory T cells (Tregs) in newborn mice. In addition, P. UF1 protected mice from intestinal Listeria monocytogenes (L. m) infection. P. UF1 also functionally sustained the gut microbiota and induced critical B vitamin metabolites implicated in the regulation of T cell immunity during L. m intestinal infection. Transcriptomic analysis of P. UF1-induced Th17 cells revealed genes involved in the differentiation and regulation of these cells. These results illustrate the potency of P. UF1 in the enhancement of neonatal host defense against intestinal pathogen infection.
Collapse
|
64
|
Oliver E, Pope C, Clarke E, Langton Hewer C, Ogunniyi AD, Paton JC, Mitchell T, Malley R, Finn A. Th17 responses to pneumococcus in blood and adenoidal cells in children. Clin Exp Immunol 2019; 195:213-225. [PMID: 30325010 PMCID: PMC6330644 DOI: 10.1111/cei.13225] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2018] [Indexed: 11/30/2022] Open
Abstract
Pneumococcal infections cause a large global health burden, and the search for serotype-independent vaccines continues. Existing conjugate vaccines reduce nasopharyngeal colonization by target serotypes. Such mucosal effects of novel antigens may similarly be important. CD4+ Th17 cell-dependent, antibody-independent reductions in colonization and enhanced clearance have been described in mice. Here we describe the evaluation of T helper type 17 (Th17) cytokine responses to candidate pneumococcal protein vaccine antigens in human cell culture, using adenoidal and peripheral blood mononuclear cells. Optimal detection of interleukin (IL)-17A was at day 7, and of IL-22 at day 11, in these primary cell cultures. Removal of CD45RO+ memory T cells abolished these responses. Age-associated increases in magnitude of responses were evident for IL-17A, but not IL-22, in adenoidal cells. There was a strong correlation between individual IL-17A and IL-22 responses after pneumococcal antigen stimulation (P < 0·015). Intracellular cytokine staining following phorbol myristate acetate (PMA)/ionomycin stimulation demonstrated that > 30% CD4+ T cells positive for IL-22 express the innate markers γδT cell receptor and/or CD56, with much lower proportions for IL-17A+ cells (P < 0·001). Responses to several vaccine candidate antigens were observed but were consistently absent, particularly in blood, to PhtD (P < 0·0001), an antigen recently shown not to impact colonization in a clinical trial of a PhtD-containing conjugate vaccine in infants. The data presented and approach discussed have the potential to assist in the identification of novel vaccine antigens aimed at reducing pneumococcal carriage and transmission, thus improving the design of empirical clinical trials.
Collapse
Affiliation(s)
- E. Oliver
- School of Cellular and Molecular Medicine, Biomedical Sciences BuildingUniversity of BristolBristolUK
| | - C. Pope
- School of Cellular and Molecular Medicine, Biomedical Sciences BuildingUniversity of BristolBristolUK
| | - E. Clarke
- Vaccines and Immunity Theme, MRC Unit The GambiaFajaraThe Gambia
| | | | - A. D. Ogunniyi
- Australian Centre for Antimicrobial Resistance Ecology, School of Animal and Veterinary SciencesThe University of AdelaideAdelaideAustralia
| | - J. C. Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical ScienceUniversity of AdelaideAdelaideAustralia
| | - T. Mitchell
- Institute of Microbiology and InfectionUniversity of BirminghamBirminghamUK
| | - R. Malley
- Division of Infectious Diseases, Department of MedicineChildren’s Hospital and Harvard Medical SchoolBostonMAUSA
| | - A. Finn
- School of Cellular and Molecular Medicine, Biomedical Sciences BuildingUniversity of BristolBristolUK
| |
Collapse
|
65
|
Ardain A, Porterfield JZ, Kløverpris HN, Leslie A. Type 3 ILCs in Lung Disease. Front Immunol 2019; 10:92. [PMID: 30761149 PMCID: PMC6361816 DOI: 10.3389/fimmu.2019.00092] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/14/2019] [Indexed: 12/12/2022] Open
Abstract
The lungs represent a complex immune setting, balancing external environmental signals with a poised immune response that must protect from infection, mediate tissue repair, and maintain lung function. Innate lymphoid cells (ILCs) play a central role in tissue repair and homeostasis, and mediate protective immunity in a variety of mucosal tissues, including the lung. All three ILC subsets are present in the airways of both mice and humans; and ILC2s shown to have pivotal roles in asthma, airway hyper-responsiveness, and parasitic worm infection. The involvement of ILC3s in respiratory diseases is less well-defined, but they are known to be critical in homeostasis, infection and inflammation at other mucosal barriers, such as the gut. Moreover, they are important players in the IL17/IL22 axis, which is key to lung health. In this review, we discuss the emerging role of ILC3s in the context of infectious and inflammatory lung diseases, with a focus on data from human subjects.
Collapse
Affiliation(s)
- Amanda Ardain
- Africa Health Research Institute, Durban, South Africa
- College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - James Zachary Porterfield
- Africa Health Research Institute, Durban, South Africa
- College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Yale School of Public Health, Yale University, New Haven, CT, United States
| | - Henrik N. Kløverpris
- Africa Health Research Institute, Durban, South Africa
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infection and Immunity, University College London, London, United Kingdom
| | - Alasdair Leslie
- Africa Health Research Institute, Durban, South Africa
- Department of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|
66
|
Snyder LM, McDaniel KL, Tian Y, Wei CH, Kennett MJ, Patterson AD, Ross AC, Cantorna MT. Retinoic Acid Mediated Clearance of Citrobacter rodentium in Vitamin A Deficient Mice Requires CD11b+ and T Cells. Front Immunol 2019; 9:3090. [PMID: 30671060 PMCID: PMC6331472 DOI: 10.3389/fimmu.2018.03090] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 12/13/2018] [Indexed: 12/27/2022] Open
Abstract
Vitamin A deficiency affects over 250 million preschool-age children worldwide and is associated with increased childhood mortality and risk of developing enteric infections. Vitamin A deficient (A-) mice developed chronic Citrobacter rodentium infection. A single oral dose of retinoic acid (RA) at d7 post-infection was sufficient to induce clearance of the pathogen in A- mice. RA treatment of A- mice induced il17 expression in the colon. In A- mice, colonic IL-17 was primarily produced by CD11b+ cells; however, in A+ mice, the major source of colonic IL-17 was CD4+ T cells. To determine the cellular targets of vitamin A required for host resistance to C. rodentium, mice that express a dominant negative (dn) retinoic acid receptor (RAR) in T cells (T-dnRAR) or macrophage/neutrophils (LysM-dnRAR) were used. T-dnRAR mice had T cells that produced a robust intestinal IL-17 response and for 40% of the mice was enough to clear the infection. The remainder of the T-dnRAR mice developed a chronic infection. A- LysM-dnRAR mice developed early lethal infections with surviving mice becoming chronically infected. RA treatment of A- LysM-dnRAR mice was ineffective for inducing colonic IL-17 or clearing C. rodentium. Retinoid signaling is required in T cells and CD11b+ cells for complete elimination of enteric pathogens.
Collapse
Affiliation(s)
- Lindsay M Snyder
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Kaitlin L McDaniel
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Yuan Tian
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, United States.,CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Centre for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, University of Chinese Academy of Sciences, Wuhan, China
| | - Cheng-Hsin Wei
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Mary J Kennett
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, United States
| | - A Catharine Ross
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Margherita T Cantorna
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, United States
| |
Collapse
|
67
|
Shen P, Lees JA, Bee GCW, Brown SP, Weiser JN. Pneumococcal quorum sensing drives an asymmetric owner-intruder competitive strategy during carriage via the competence regulon. Nat Microbiol 2018; 4:198-208. [PMID: 30546100 DOI: 10.1038/s41564-018-0314-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 10/30/2018] [Indexed: 11/09/2022]
Abstract
Competition among microorganisms is a key determinant of successful host colonization and persistence. For Streptococcus pneumoniae, lower than predicted rates of co-colonizing strains suggest a competitive advantage for resident bacteria over newcomers. In light of evolutionary theory, we hypothesized that S. pneumoniae use owner-intruder asymmetries to settle contests, leading to the disproportionate success of the initial resident 'owner', regardless of the genetic identity of the 'intruder'. We investigated the determinants of within-host competitive success utilizing S. pneumoniae colonization of the upper respiratory tract of infant mice. Within 6 h, colonization by the resident inhibited colonization by an isogenic challenger. The competitive advantage of the resident was dependent on quorum sensing via the competence (Com) regulon and downstream choline binding protein D (CbpD) and on the competence-induced bacteriocins A and B (CibAB) implicated in fratricide. CbpD and CibAB are highly conserved across pneumococcal lineages, indicating evolutionary advantages for asymmetric competitive strategies within the species. Mathematical modelling supported a significant role for quorum sensing via the Com regulon in competition, even for strains with different competitive advantages. Our study suggests that asymmetric owner-intruder competitive strategies do not require complex cognition and are used by a major human pathogen to determine 'ownership' of human hosts.
Collapse
Affiliation(s)
- Pamela Shen
- Department of Microbiology, New York University School of Medicine, New York, NY, USA
| | - John A Lees
- Department of Microbiology, New York University School of Medicine, New York, NY, USA
| | - Gavyn Chern Wei Bee
- Department of Microbiology, New York University School of Medicine, New York, NY, USA
| | - Sam P Brown
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jeffrey N Weiser
- Department of Microbiology, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
68
|
Jochems SP, Marcon F, Carniel BF, Holloway M, Mitsi E, Smith E, Gritzfeld JF, Solórzano C, Reiné J, Pojar S, Nikolaou E, German EL, Hyder-Wright A, Hill H, Hales C, de Steenhuijsen Piters WAA, Bogaert D, Adler H, Zaidi S, Connor V, Gordon SB, Rylance J, Nakaya HI, Ferreira DM. Inflammation induced by influenza virus impairs human innate immune control of pneumococcus. Nat Immunol 2018; 19:1299-1308. [PMID: 30374129 PMCID: PMC6241853 DOI: 10.1038/s41590-018-0231-y] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/07/2018] [Indexed: 12/02/2022]
Abstract
Colonization of the upper respiratory tract by pneumococcus is important both as a determinant of disease and for transmission into the population. The immunological mechanisms that contain pneumococcus during colonization are well studied in mice but remain unclear in humans. Loss of this control of pneumococcus following infection with influenza virus is associated with secondary bacterial pneumonia. We used a human challenge model with type 6B pneumococcus to show that acquisition of pneumococcus induced early degranulation of resident neutrophils and recruitment of monocytes to the nose. Monocyte function was associated with the clearance of pneumococcus. Prior nasal infection with live attenuated influenza virus induced inflammation, impaired innate immune function and altered genome-wide nasal gene responses to the carriage of pneumococcus. Levels of the cytokine CXCL10, promoted by viral infection, at the time pneumococcus was encountered were positively associated with bacterial load.
Collapse
Affiliation(s)
- Simon P Jochems
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK.
| | - Fernando Marcon
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paolo, Brazil
| | - Beatriz F Carniel
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Mark Holloway
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Elena Mitsi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Emma Smith
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Jenna F Gritzfeld
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Carla Solórzano
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Jesús Reiné
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Sherin Pojar
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Elissavet Nikolaou
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Esther L German
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Angie Hyder-Wright
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
- Royal Liverpool and Broadgreen University Hospital, Liverpool, UK
| | - Helen Hill
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
- Royal Liverpool and Broadgreen University Hospital, Liverpool, UK
| | - Caz Hales
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
- Royal Liverpool and Broadgreen University Hospital, Liverpool, UK
| | - Wouter A A de Steenhuijsen Piters
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
- Department of Paediatric Immunology and Infectious Diseases, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Debby Bogaert
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
- Department of Paediatric Immunology and Infectious Diseases, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Hugh Adler
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Seher Zaidi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Victoria Connor
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
- Royal Liverpool and Broadgreen University Hospital, Liverpool, UK
| | - Stephen B Gordon
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - Jamie Rylance
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Helder I Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paolo, Brazil.
| | - Daniela M Ferreira
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK.
| |
Collapse
|
69
|
Huang CC, Wu PW, Chen CL, Wang CH, Lee TJ, Tsai CN, Chiu CH. IL-17A expression in the adenoid tissue from children with sleep disordered breathing and its association with pneumococcal carriage. Sci Rep 2018; 8:16770. [PMID: 30425273 PMCID: PMC6233154 DOI: 10.1038/s41598-018-35169-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 08/20/2018] [Indexed: 12/13/2022] Open
Abstract
Tonsil and adenoid-tissue hypertrophy (AH) is the most common cause of pediatric sleep-disordered breathing (SDB), with AH possibly initiated by repeated exposure to infectious agents or allergens. Here, we evaluated IL-17A activity in adenoid tissue from children with SDB and its association with AH and pneumococcal carriage. Thirty-five children (aged 3-12 years) with SDB and receiving adenoidectomy and tonsillectomy were enrolled. During surgery, nasopharyngeal carriage was determined by bacterial culture and multiplex PCR via nasopharyngeal swab, and adenoid samples were collected. IL-17A and associated cytokine expression was evaluated by real-time PCR and western blotting. The mRNA analysis showed that IL-17A level, IL-17A:IL-10 ratio, and RAR-related orphan receptor-γt:forkhead box P3 ratio were significantly higher in adenoid tissues with AH, as were IL-17A level and IL-17A:IL-10 ratio in adenoid tissues with pneumococcal carriage. Additionally, pneumococcal carriage was more common in nasopharyngeal adenoids from patients without AH than those with AH. IL-17A was upregulated in adenoid tissues from patients with AH and with pneumococcal carriage. These results suggested that pneumococcal carriage initiates an IL-17A-mediated immune response in nasopharyngeal adenoids, which might be associated with AH in patients with SDB.
Collapse
Affiliation(s)
- Chien-Chia Huang
- Division of Rhinology, Department of Otolaryngology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Pei-Wen Wu
- Division of Rhinology, Department of Otolaryngology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan.,Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Keelung, Taiwan
| | - Chyi-Liang Chen
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chun-Hua Wang
- Department of Thoracic Medicine, Chang Gung Memorial Hospital and Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Ta-Jen Lee
- Division of Rhinology, Department of Otolaryngology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Chi-Neu Tsai
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Cheng-Hsun Chiu
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan. .,Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
70
|
Lu YJ, Oliver E, Zhang F, Pope C, Finn A, Malley R. Screening for Th17-Dependent Pneumococcal Vaccine Antigens: Comparison of Murine and Human Cellular Immune Responses. Infect Immun 2018; 86:e00490-18. [PMID: 30150255 PMCID: PMC6204694 DOI: 10.1128/iai.00490-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 08/17/2018] [Indexed: 12/14/2022] Open
Abstract
Conjugate vaccines against Streptococcus pneumoniae have significantly reduced the incidence of diseases caused by the serotypes included in those vaccines; however, there is still a need for vaccines that confer serotype-independent protection. In the current study, we have constructed a library of conserved surface proteins from S. pneumoniae and have screened for IL-17A and IL-22 production in human immune cells obtained from adenoidal/tonsillar tissues of children and IL-17A production in splenocytes from mice that had been immunized with a killed whole-cell vaccine or previously exposed to pneumococcus. A positive correlation was found between the rankings of proteins from human IL-17A and IL-22 screens, but not between those from human and mouse screens. All proteins were tested for protection against colonization, and we identified protective antigens that are IL-17A dependent. We found that the likelihood of finding a protective antigen is significantly higher for groups of proteins ranked in the top 50% of all three screens than for groups of proteins ranked in the bottom 50% of all three. The results thus confirmed the value of such screens for identifying Th17 antigens. Further, these experiments have evaluated and compared the breadth of human and mouse Th17 responses to pneumococcal colonization and have enabled the identification of potential vaccine candidates based on immunological responses in mouse and human cells.
Collapse
Affiliation(s)
- Ying-Jie Lu
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Elizabeth Oliver
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Fan Zhang
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Caroline Pope
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Adam Finn
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
- School of Population Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Richard Malley
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
71
|
Age-related differences in IL-1 signaling and capsule serotype affect persistence of Streptococcus pneumoniae colonization. PLoS Pathog 2018; 14:e1007396. [PMID: 30379943 PMCID: PMC6231672 DOI: 10.1371/journal.ppat.1007396] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 11/12/2018] [Accepted: 10/10/2018] [Indexed: 12/20/2022] Open
Abstract
Young age is a risk factor for prolonged colonization by common pathogens residing in their upper respiratory tract (URT). Why children present with more persistent colonization is unknown and there is relatively little insight into the host-pathogen interactions that contribute to persistent colonization. To identify factors permissive for persistent colonization during infancy, we utilized an infant mouse model of Streptococcus pneumoniae colonization in which clearance from the mucosal surface of the URT requires many weeks to months. Loss of a single bacterial factor, the pore-forming toxin pneumolysin (Ply), and loss of a single host factor, IL-1α, led to more persistent colonization. Exogenous administration of Ply promoted IL-1 responses and clearance, and intranasal treatment with IL-1α was sufficient to reduce colonization density. Major factors known to affect the duration of natural colonization include host age and pneumococcal capsular serotype. qRT-PCR analysis of the uninfected URT mucosa showed reduced baseline expression of genes involved in IL-1 signaling in infant compared to adult mice. In line with this observation, IL-1 signaling was important in initiating clearance in adult mice but had no effect on early colonization of infant mice. In contrast to the effect of age, isogenic constructs of different capsular serotype showed differences in colonization persistence but induced similar IL-1 responses. Altogether, this work underscores the importance of toxin-induced IL-1α responses in determining the outcome of colonization, clearance versus persistence. Our findings about IL-1 signaling as a function of host age may provide an explanation for the increased susceptibility and more prolonged colonization during early childhood. During early childhood, opportunistic pathogens are often carried in the upper respiratory tract (URT) for prolonged periods of time. Why young children experience more persistent carriage is unclear and there is little understanding of host-bacteria interactions that affect persistence, especially in infants. Here, we utilized an infant mouse model of Streptococcus pneumoniae colonization, a common pathogen of the infant URT, that persists for several months. We identified that clearance is dictated by bacterial expression of a single pneumococcal toxin, pneumolysin, and by the host response via a single cytokine, IL-1α, that activates IL-1 signaling. Absence of either of these factors led to increased persistence of S. pneumoniae. We discovered that the infant URT shows repression of IL-1 signaling compared to adults. Our study presents new insight into the importance of IL-1 signaling in clearance of persistent URT carriage and may provide an explanation why infants present with more persistent carriage by common URT pathogens.
Collapse
|
72
|
A recombinant conjugated pneumococcal vaccine that protects against murine infections with a similar efficacy to Prevnar-13. NPJ Vaccines 2018; 3:53. [PMID: 30393571 PMCID: PMC6208403 DOI: 10.1038/s41541-018-0090-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 10/08/2018] [Indexed: 11/09/2022] Open
Abstract
The pneumococcal conjugate vaccine (PCV) strongly protects against vaccine serotypes, but the rapid expansion of non-vaccine serotype disease and the vaccine's high expense has reduced its overall impact. We have developed Protein Glycan Coupling Technology (PGCT) as a flexible methodology for making low-cost polysaccharide/protein glycoconjugates recombinantly in Escherichia coli. We have used PGCT to make a recombinant PCV containing serotype 4 capsular polysaccharide linked to the Streptococcus pneumoniae proteins NanA, PiuA, and Sp0148. The introduction of the Campylobacter jejuni UDP-glucose 4-epimerase gene GalE (gne) into E. coli improved the yield of the resulting glycoprotein. PGCT glycoconjugate vaccination generated strong antibody responses in mice to both the capsule and the carrier protein antigens, with the PiuA/capsule glycoconjugate inducing similar anti-capsular antibody responses as the commercial PCV Prevnar-13. Antibody responses to PGCT glycoconjugates opsonised S. pneumoniae and Streptococcus mitis expressing the serotype 4 capsule and promoted neutrophil phagocytosis of S. pneumoniae to a similar level as antisera generated by vaccination with Prevnar-13. Vaccination with the PGCT glycoconjugates protected mice against meningitis and septicaemia with the same efficacy as vaccination with Prevnar-13. In addition, vaccination with the protein antigen components from PGCT glycoconjugates alone provided partial protection against septicaemia and colonisation. These data demonstrate that a vaccine made by PGCT is as effective as Prevnar-13, identifies PiuA as a carrier protein for glycoconjugate vaccines, and demonstrates that linking capsular antigen to S. pneumoniae protein antigens has additional protective benefits that could provide a degree of serotype-independent immunity.
Collapse
|
73
|
Protection against Staphylococcus aureus Colonization and Infection by B- and T-Cell-Mediated Mechanisms. mBio 2018; 9:mBio.01949-18. [PMID: 30327437 PMCID: PMC6191547 DOI: 10.1128/mbio.01949-18] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
S. aureus is a leading cause of healthcare- and community-associated bacterial infections. S. aureus causes various illnesses, including bacteremia, meningitis, endocarditis, pneumonia, osteomyelitis, sepsis, and skin and soft tissue infections. S. aureus colonizes between 20 and 80% of humans; carriers are at increased risk for infection and transmission to others. The spread of multidrug-resistant strains limits antibiotic treatment options. Vaccine development against S. aureus has been unsuccessful to date, likely due to an inadequate understanding about the mechanisms of immune defense against this pathogen. The significance of our work is in illustrating the necessity of generating multipronged B-cell, Th1-, and Th17-mediated responses to S. aureus antigens in conferring enhanced and broad protection against S. aureus invasive infection, skin and soft tissue infection, and mucosal colonization. Our work thus, provides important insights for future vaccine development against this pathogen. Staphylococcus aureus is a major cause of morbidity and mortality worldwide. S. aureus colonizes 20 to 80% of humans at any one time and causes a variety of illnesses. Strains that are resistant to common antibiotics further complicate management. S. aureus vaccine development has been unsuccessful so far, largely due to the incomplete understanding of the mechanisms of protection against this pathogen. Here, we studied the role of different aspects of adaptive immunity induced by an S. aureus vaccine in protection against S. aureus bacteremia, dermonecrosis, skin abscess, and gastrointestinal (GI) colonization. We show that, depending on the challenge model, the contributions of vaccine-induced S. aureus-specific antibody and Th1 and Th17 responses to protection are different: antibodies play a major role in reducing mortality during S. aureus bacteremia, whereas Th1 or Th17 responses are essential for prevention of S. aureus skin abscesses and the clearance of bacteria from the GI tract. Both antibody- and T-cell-mediated mechanisms contribute to prevention of S. aureus dermonecrosis. Engagement of all three immune pathways results in the most robust protection under each pathological condition. Therefore, our results suggest that eliciting multipronged humoral and cellular responses to S. aureus antigens may be critical to achieve effective and comprehensive immune defense against this pathogen.
Collapse
|
74
|
Loughran ST, Power PA, Maguire PT, McQuaid SL, Buchanan PJ, Jonsdottir I, Newman RW, Harvey R, Johnson PA. Influenza infection directly alters innate IL-23 and IL-12p70 and subsequent IL-17A and IFN-γ responses to pneumococcus in vitro in human monocytes. PLoS One 2018; 13:e0203521. [PMID: 30192848 PMCID: PMC6128554 DOI: 10.1371/journal.pone.0203521] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 08/22/2018] [Indexed: 12/21/2022] Open
Abstract
IMPORTANCE Influenza virus is highly contagious and poses substantial public health problems due to its strong association with morbidity and mortality. Approximately 250,000-500,000 deaths are caused by seasonal influenza virus annually, and this figure increases during periods of pandemic infections. Most of these deaths are due to secondary bacterial pneumonia. Influenza-bacterial superinfection can result in hospitalisation and/or death of both patients with pre-existing lung disease or previously healthy individuals. The importance of our research is in determining that influenza and its component haemagglutinin has a direct effect on the classic pneumococcus induced pathways to IL-17A in our human ex vivo model. Our understanding of the mechanism which leaves people exposed to influenza infection during superinfection remain unresolved. This paper demonstrates that early infection of monocytes inhibits an arm of immunity crucial to bacterial clearance. Understanding this mechanism may provide alternative interventions in the case of superinfection with antimicrobial resistant strains of bacteria.
Collapse
Affiliation(s)
- Sinead T. Loughran
- Viral Immunology Laboratory, School of Nursing and Human Sciences, Dublin City University, Dublin, Ireland
| | - Patrick A. Power
- Viral Immunology Laboratory, School of Nursing and Human Sciences, Dublin City University, Dublin, Ireland
| | - Paula T. Maguire
- Viral Immunology Laboratory, School of Nursing and Human Sciences, Dublin City University, Dublin, Ireland
| | - Samantha L. McQuaid
- Viral Immunology Laboratory, School of Nursing and Human Sciences, Dublin City University, Dublin, Ireland
| | - Paul J. Buchanan
- Translational Cancer Physiology Laboratory, School of Nursing and Human Sciences, Dublin City University, Dublin, Ireland
| | - Ingileif Jonsdottir
- Department of Immunology, Landspitali, The National University Hospital of Iceland, Reykjavik, Iceland
| | - Robert W. Newman
- National Institute for Biological Standards and Controls, Potters Bar, Herts, United Kingdom
| | - Ruth Harvey
- National Institute for Biological Standards and Controls, Potters Bar, Herts, United Kingdom
| | - Patricia A. Johnson
- Viral Immunology Laboratory, School of Nursing and Human Sciences, Dublin City University, Dublin, Ireland
| |
Collapse
|
75
|
González-Miró M, Radecker AM, Rodríguez-Noda LM, Fariñas-Medina M, Zayas-Vignier C, Hernández-Cedeño M, Serrano Y, Cardoso F, Santana-Mederos D, García-Rivera D, Valdés-Balbín Y, Vérez-Bencomo V, Rehm BHA. Design and Biological Assembly of Polyester Beads Displaying Pneumococcal Antigens as Particulate Vaccine. ACS Biomater Sci Eng 2018; 4:3413-3424. [DOI: 10.1021/acsbiomaterials.8b00579] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Majela González-Miró
- Finlay Vaccine Institute, 27th Avenue, No. 19805 between 198 and 202, La Lisa, Havana 11600, Cuba
- Institute of Fundamental Sciences, Massey University, Colombo Road, Palmerston North 4422, New Zealand
| | - Anna-Maria Radecker
- Institute of Fundamental Sciences, Massey University, Colombo Road, Palmerston North 4422, New Zealand
| | - Laura M. Rodríguez-Noda
- Finlay Vaccine Institute, 27th Avenue, No. 19805 between 198 and 202, La Lisa, Havana 11600, Cuba
| | - Mildrey Fariñas-Medina
- Finlay Vaccine Institute, 27th Avenue, No. 19805 between 198 and 202, La Lisa, Havana 11600, Cuba
| | - Caridad Zayas-Vignier
- Finlay Vaccine Institute, 27th Avenue, No. 19805 between 198 and 202, La Lisa, Havana 11600, Cuba
| | - Mabel Hernández-Cedeño
- Finlay Vaccine Institute, 27th Avenue, No. 19805 between 198 and 202, La Lisa, Havana 11600, Cuba
| | - Yohana Serrano
- Finlay Vaccine Institute, 27th Avenue, No. 19805 between 198 and 202, La Lisa, Havana 11600, Cuba
| | - Félix Cardoso
- Finlay Vaccine Institute, 27th Avenue, No. 19805 between 198 and 202, La Lisa, Havana 11600, Cuba
| | - Darielys Santana-Mederos
- Finlay Vaccine Institute, 27th Avenue, No. 19805 between 198 and 202, La Lisa, Havana 11600, Cuba
| | - Dagmar García-Rivera
- Finlay Vaccine Institute, 27th Avenue, No. 19805 between 198 and 202, La Lisa, Havana 11600, Cuba
| | - Yury Valdés-Balbín
- Finlay Vaccine Institute, 27th Avenue, No. 19805 between 198 and 202, La Lisa, Havana 11600, Cuba
| | - Vicente Vérez-Bencomo
- Finlay Vaccine Institute, 27th Avenue, No. 19805 between 198 and 202, La Lisa, Havana 11600, Cuba
| | - Bernd H. A. Rehm
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan Campus, Nathan, Queensland 4111, Australia
| |
Collapse
|
76
|
Enzymatic Hydrolysis of Pneumococcal Capsular Polysaccharide Renders the Bacterium Vulnerable to Host Defense. Infect Immun 2018; 86:IAI.00316-18. [PMID: 29866907 DOI: 10.1128/iai.00316-18] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 05/30/2018] [Indexed: 12/15/2022] Open
Abstract
Despite a century of investigation, Streptococcus pneumoniae remains a major human pathogen, causing a number of diseases, such as pneumonia, meningitis, and otitis media. Like many encapsulated pathogens, the capsular polysaccharide (CPS) of S. pneumoniae is a critical component for colonization and virulence in mammalian hosts. This study aimed to evaluate the protective role of a glycoside hydrolase, Pn3Pase, targeting the CPS of type 3 S. pneumoniae, which is one of the most virulent serotypes. We have assessed the ability of Pn3Pase to degrade the capsule on a live type 3 strain. Through in vitro assays, we observed that Pn3Pase treatment increases the bacterium's susceptibility to phagocytosis by macrophages and complement-mediated killing by neutrophils. We have demonstrated that in vivo Pn3Pase treatment reduces nasopharyngeal colonization and protects mice from sepsis caused by type 3 S. pneumoniae Due to the increasing shifts in serotype distribution, the rise in drug-resistant strains, and poor immune responses to vaccine-included serotypes, it is necessary to investigate approaches to combat pneumococcal infections. This study evaluates the interaction of pneumococcal CPS with the host at molecular, cellular, and systemic levels and offers an alternative therapeutic approach for diseases caused by S. pneumoniae through enzymatic hydrolysis of the CPS.
Collapse
|
77
|
Pichichero ME. Pneumococcal whole-cell and protein-based vaccines: changing the paradigm. Expert Rev Vaccines 2018; 16:1181-1190. [PMID: 29130395 DOI: 10.1080/14760584.2017.1393335] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Epidemiologic evaluations of Streptococcus pneumoniae nasopharyngeal (NP) colonization and pneumococcal disease suggest that newer serotypes in future formulations of pneumococcal conjugate vaccines (PCVs) are needed and there may need to be continued reformulations because there are many new emerging serotypes expressed by pneumococci. Areas covered: Mechanisms of protection by next-generation whole-cell vaccine (WCV) and/or multi-component pneumococcal purified protein vaccines (PPVs) in development for prevention of pneumococcal infections. Expert commentary: A long-term strategy for prevention of pneumococcal disease will likely include WCV and PPVs. However these vaccines will impact disease pathogenesis in a different manner than PCVs. Prevention of pneumococcal NP colonization should not be expected, nor is it desirable because risks for NP colonization by other replacement organisms into the ecological niche vacated by all pneumococci may have consequences. The expression biology of capsule and surface protein antigens are phase dependent. Therefore, the immune response will be different and the mechanism of protection divergent. WCVs and PPVs may be alternative strategies in low income developing countries to protect against invasive disease and reduce NP carriage load.
Collapse
Affiliation(s)
- Michael E Pichichero
- a Rochester General Hospital Research Institute , Center for Infectious Diseases and Immunology , Rochester , NY , USA
| |
Collapse
|
78
|
Weiser JN, Ferreira DM, Paton JC. Streptococcus pneumoniae: transmission, colonization and invasion. Nat Rev Microbiol 2018; 16:355-367. [PMID: 29599457 PMCID: PMC5949087 DOI: 10.1038/s41579-018-0001-8] [Citation(s) in RCA: 560] [Impact Index Per Article: 93.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Streptococcus pneumoniae has a complex relationship with its obligate human host. On the one hand, the pneumococci are highly adapted commensals, and their main reservoir on the mucosal surface of the upper airways of carriers enables transmission. On the other hand, they can cause severe disease when bacterial and host factors allow them to invade essentially sterile sites, such as the middle ear spaces, lungs, bloodstream and meninges. Transmission, colonization and invasion depend on the remarkable ability of S. pneumoniae to evade or take advantage of the host inflammatory and immune responses. The different stages of pneumococcal carriage and disease have been investigated in detail in animal models and, more recently, in experimental human infection. Furthermore, widespread vaccination and the resulting immune pressure have shed light on pneumococcal population dynamics and pathogenesis. Here, we review the mechanistic insights provided by these studies on the multiple and varied interactions of the pneumococcus and its host.
Collapse
|
79
|
Ritchie ND, Ritchie R, Bayes HK, Mitchell TJ, Evans TJ. IL-17 can be protective or deleterious in murine pneumococcal pneumonia. PLoS Pathog 2018; 14:e1007099. [PMID: 29813133 PMCID: PMC5993294 DOI: 10.1371/journal.ppat.1007099] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/08/2018] [Accepted: 05/14/2018] [Indexed: 12/21/2022] Open
Abstract
Streptococcus pneumoniae is the major bacterial cause of community-acquired pneumonia, and the leading agent of childhood pneumonia deaths worldwide. Nasal colonization is an essential step prior to infection. The cytokine IL-17 protects against such colonization and vaccines that enhance IL-17 responses to pneumococcal colonization are being developed. The role of IL-17 in host defence against pneumonia is not known. To address this issue, we have utilized a murine model of pneumococcal pneumonia in which the gene for the IL-17 cytokine family receptor, Il17ra, has been inactivated. Using this model, we show that IL-17 produced predominantly from γδ T cells protects mice against death from the invasive TIGR4 strain (serotype 4) which expresses a relatively thin capsule. However, in pneumonia produced by two heavily encapsulated strains with low invasive potential (serotypes 3 and 6B), IL-17 significantly enhanced mortality. Neutrophil uptake and killing of the serotype 3 strain was significantly impaired compared to the serotype 4 strain and depletion of neutrophils with antibody enhanced survival of mice infected with the highly encapsulated SRL1 strain. These data strongly suggest that IL-17 mediated neutrophil recruitment to the lungs clears infection from the invasive TIGR4 strain but that lung neutrophils exacerbate disease caused by the highly encapsulated pneumococcal strains. Thus, whilst augmenting IL-17 immune responses against pneumococci may decrease nasal colonization, this may worsen outcome during pneumonia caused by some strains.
Collapse
MESH Headings
- Animals
- Bacteremia/immunology
- Bacteremia/microbiology
- Bacterial Capsules/immunology
- Bacterial Capsules/ultrastructure
- Bronchoalveolar Lavage Fluid/cytology
- Bronchoalveolar Lavage Fluid/microbiology
- Disease Models, Animal
- Interleukin-17/immunology
- Lung/cytology
- Lung/enzymology
- Lung/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Electron, Transmission
- Microscopy, Fluorescence
- Nasopharynx/microbiology
- Neutrophils/cytology
- Neutrophils/immunology
- Peroxidase/metabolism
- Phagocytosis
- Pneumonia, Pneumococcal/immunology
- Pneumonia, Pneumococcal/mortality
- Pneumonia, Pneumococcal/prevention & control
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Interleukin-17/genetics
- Specific Pathogen-Free Organisms
- Streptococcus pneumoniae/immunology
- Streptococcus pneumoniae/ultrastructure
Collapse
Affiliation(s)
- Neil D. Ritchie
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Ryan Ritchie
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Hannah K. Bayes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Tim J. Mitchell
- Institute of Microbiology and Infection, College of Medical and Dental Sciences University of Birmingham, Birmingham, United Kingdom
| | - Tom J. Evans
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
80
|
Evaluation of the Role of stat3 in Antibody and T H17-Mediated Responses to Pneumococcal Immunization and Infection by Use of a Mouse Model of Autosomal Dominant Hyper-IgE Syndrome. Infect Immun 2018; 86:IAI.00024-18. [PMID: 29463618 DOI: 10.1128/iai.00024-18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 02/13/2018] [Indexed: 12/12/2022] Open
Abstract
Loss-of-function mutations in the signal transducer and activator of transcription 3 gene (stat3) result in autosomal dominant hyper-IgE syndrome (AD-HIES), a condition in which patients have recurrent debilitating infections, including frequent pneumococcal and staphylococcal pneumonias. stat3 mutations cause defective adaptive TH17 cellular responses, an immune mechanism believed to be critical for clearance of pneumococcal colonization and diminished antibody responses. Here we wished to evaluate the role of stat3 in the clearance of pneumococcal carriage and immunity using mice with a stat3 mutation recapitulating AD-HIES. We show here that naive AD-HIES mice have prolonged nasal carriage of pneumococcus compared to WT mice. Mutant and wild-type mice were then immunized with a pneumococcal whole-cell vaccine (WCV) that provides TH17-mediated protection against pneumococcal colonization and antibody-mediated protection against pneumonia and sepsis. WCV-immunized AD-HIES mice made significantly less pneumococcus-specific interleukin-17A (IL-17A) and antibody than WT mice. The WCV-elicited protection against colonization was abrogated in AD-HIES mice, but immunization with WCV still protected AD-HIES mice against aspiration pneumonia/sepsis. Taken together, our results suggest that impaired clearance of nasopharyngeal carriage due to poor adaptive IL-17A responses may contribute to the increased rates of pneumococcal respiratory infection in AD-HIES patients.
Collapse
|
81
|
van Ravenhorst MB, den Hartog G, van der Klis FRM, van Rooijen DM, Sanders EAM, Berbers GAM. Induction of salivary antibody levels in Dutch adolescents after immunization with monovalent meningococcal serogroup C or quadrivalent meningococcal serogroup A, C, W and Y conjugate vaccine. PLoS One 2018; 13:e0191261. [PMID: 29672552 PMCID: PMC5908077 DOI: 10.1371/journal.pone.0191261] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/22/2017] [Indexed: 11/29/2022] Open
Abstract
Background Meningococcal infection starts with colonisation of the upper respiratory tract. Mucosal immunity is important for protection against acquisition and subsequent meningococcal carriage. In this study, we assessed salivary antibody levels against meningococcal serogroup A (MenA), W (MenW) and Y (MenY) after vaccination with a quadrivalent MenACWY conjugated vaccine. We also compared salivary meningococcal serogroup C (MenC) antibody levels after monovalent MenC and quadrivalent MenACWY conjugated vaccination. Methods Healthy participants, who had received MenC conjugate vaccine between 14 months and 3 years of age, received a (booster) MenC or MenACWY vaccination at age 10–15 years. MenA-, MenC-, MenW- and MenY-polysaccharide (PS) specific IgG and IgA levels in saliva and serum and PS specific secretory component levels in saliva were measured using the fluorescent-bead-based multiplex immunoassay. Results MenACYW vaccination increased salivary PS-specific IgA (2-fold) and IgG levels(>10-fold) for MenA, MenY, and MenW. After one year, salivary IgA levels had returned to baseline levels. Both vaccines induced an increase in salivary MenC-PS specific IgA (>3-fold) and IgG (>100-fold), with higher levels after MenC as compared to MenACWY vaccination. The antibody decay rate of MenC in saliva between one month and one year was similar for both vaccines. The overall correlation between serum and saliva IgA levels was low (R = 0.39, R = 0.58, R = 0.31, and R = 0.36 for MenA, MenC, MenW and MenY, respectively). Serogroup-PS specific IgG levels between serum and saliva correlated better (R ranged from 0.51 to 0.88). Conclusions Both primary (MenA, MenY, and MenW) and booster (MenC) parenteral meningococcal conjugate vaccination induced high salivary antibody levels. The strong correlation for MenC, MenW and MenY between saliva and serum IgG levels indicates that saliva might be used as a reliable tool to measure vaccine responses after both primary and booster meningococcal vaccination.
Collapse
Affiliation(s)
- Mariëtte B. van Ravenhorst
- CIb, National Institute of Public Health and the Environment (RIVM), Bilthoven, The Netherlands
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children’s Hospital, University Medical Center, Utrecht, The Netherlands
| | - Gerco den Hartog
- CIb, National Institute of Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | | | - Debbie M. van Rooijen
- CIb, National Institute of Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Elisabeth A. M. Sanders
- CIb, National Institute of Public Health and the Environment (RIVM), Bilthoven, The Netherlands
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children’s Hospital, University Medical Center, Utrecht, The Netherlands
| | - Guy A. M. Berbers
- CIb, National Institute of Public Health and the Environment (RIVM), Bilthoven, The Netherlands
- * E-mail:
| |
Collapse
|
82
|
Shekhar S, Khan R, Ferreira DM, Mitsi E, German E, Rørvik GH, Berild D, Schenck K, Kwon K, Petersen F. Antibodies Reactive to Commensal Streptococcus mitis Show Cross-Reactivity With Virulent Streptococcus pneumoniae Serotypes. Front Immunol 2018; 9:747. [PMID: 29713324 PMCID: PMC5911667 DOI: 10.3389/fimmu.2018.00747] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/26/2018] [Indexed: 12/05/2022] Open
Abstract
Current vaccines against Streptococcus pneumoniae, a bacterial species that afflicts people by causing a wide spectrum of diseases, do not protect against all pneumococcal serotypes. Thus, alternative vaccines to fight pneumococcal infections that target common proteins are under investigation. One promising strategy is to take advantage of immune cross-reactivity between commensal and pathogenic microbes for cross-protection. In this study, we examined the antibody-mediated cross-reactivity between S. pneumoniae and Streptococcus mitis, a commensal species closely related to S. pneumoniae. Western blot analysis showed that rabbit antisera raised against S. mitis reacted with multiple proteins of virulent S. pneumoniae strains (6B, TIGR4, and D39). Rabbit anti-S. pneumoniae IgG antibodies also showed binding to S. mitis antigens. Incubation of rabbit antisera raised against S. mitis with heterologous or homologous bacterial lysates resulted in marked inhibition of the developments of bands in the Western blots. Furthermore, plasma IgG antibodies from adult human volunteers intranasally inoculated with S. pneumoniae 6B revealed enhanced S. mitis-specific IgG titers compared with the pre-inoculation samples. Using an on-chip protein microarray representing a number of selected membrane and extracellular S. pneumoniae proteins, we identified choline-binding protein D (CbpD), cell division protein (FtsH), and manganese ABC transporter or manganese-binding adhesion lipoprotein (PsaA) as common targets of the rabbit IgG antibodies raised against S. mitis or S. pneumoniae. Cumulatively, these findings provide evidence on the antibody-mediated cross-reactivity of proteins from S. mitis and S. pneumoniae, which may have implications for development of effective and wide-range pneumococcal vaccines.
Collapse
Affiliation(s)
- Sudhanshu Shekhar
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Rabia Khan
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Daniela M Ferreira
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Elena Mitsi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Esther German
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | | | - Dag Berild
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Karl Schenck
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Keehwan Kwon
- Infectious Diseases Group, J. Craig Venter Institute, Rockville, MD, United States
| | - Fernanda Petersen
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| |
Collapse
|
83
|
Bou Ghanem EN, Maung NHT, Siwapornchai N, Goodwin AE, Clark S, Muñoz-Elías EJ, Camilli A, Gerstein RM, Leong JM. Nasopharyngeal Exposure to Streptococcus pneumoniae Induces Extended Age-Dependent Protection against Pulmonary Infection Mediated by Antibodies and CD138 + Cells. THE JOURNAL OF IMMUNOLOGY 2018; 200:3739-3751. [PMID: 29661828 DOI: 10.4049/jimmunol.1701065] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 03/22/2018] [Indexed: 11/19/2022]
Abstract
Streptococcus pneumoniae commonly resides asymptomatically in the nasopharyngeal (NP) cavity of healthy individuals but can cause life-threatening pulmonary and systemic infections, particularly in the elderly. NP colonization results in a robust immune response that protects against invasive infections. However, the duration, mechanism, and cellular component of such responses are poorly understood. In this study, we found that repeated NP exposure of mice to S. pneumoniae TIGR4 strain results in pneumococcal-specific Ab responses that protect against lethal lung challenge. Abs were necessary and sufficient for protection because Ab-deficient μMT mice did not develop postexposure protection, only becoming resistant to lung infection after transfer of immune sera from NP-exposed mice. T cells contributed to immunity at the time of NP exposure, but neither CD4+ nor CD8+ T cells were required. The protective activity was detectable 20 wk after exposure and was maintained in irradiated mice, suggesting involvement of long-lived Ab-secreting cells (ASC), which are radioresistant and secrete Abs for extended periods of time in the absence of T cells or persistent Ag. CD138+ bone marrow cells, likely corresponding to long-lived ASC, were sufficient to confer protection. NP exposure of aged mice failed to protect against subsequent lung infection despite eliciting a robust Ab response. Furthermore, transfer of CD138+ bone marrow cells or sera from NP-exposed old mice failed to protect naive young mice. These findings suggest that NP exposure elicits extended protection against pneumococcal lung infection by generating long-lived CD138+ ASC and that the protective efficacy of these responses declines with age.
Collapse
Affiliation(s)
- Elsa N Bou Ghanem
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY 14203
| | - Nang H Tin Maung
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655
| | - Nalat Siwapornchai
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111
| | - Aaron E Goodwin
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655
| | - Stacie Clark
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111.,Graduate Program in Molecular Microbiology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
| | | | - Andrew Camilli
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111.,Howard Hughes Medical Institute, Boston, MA 02111
| | - Rachel M Gerstein
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655
| | - John M Leong
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111;
| |
Collapse
|
84
|
Hakansson AP, Orihuela CJ, Bogaert D. Bacterial-Host Interactions: Physiology and Pathophysiology of Respiratory Infection. Physiol Rev 2018; 98:781-811. [PMID: 29488821 PMCID: PMC5966719 DOI: 10.1152/physrev.00040.2016] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 09/08/2017] [Accepted: 09/11/2017] [Indexed: 02/06/2023] Open
Abstract
It has long been thought that respiratory infections are the direct result of acquisition of pathogenic viruses or bacteria, followed by their overgrowth, dissemination, and in some instances tissue invasion. In the last decades, it has become apparent that in contrast to this classical view, the majority of microorganisms associated with respiratory infections and inflammation are actually common members of the respiratory ecosystem and only in rare circumstances do they cause disease. This suggests that a complex interplay between host, environment, and properties of colonizing microorganisms together determines disease development and its severity. To understand the pathophysiological processes that underlie respiratory infectious diseases, it is therefore necessary to understand the host-bacterial interactions occurring at mucosal surfaces, along with the microbes inhabiting them, during symbiosis. Current knowledge regarding host-bacterial interactions during asymptomatic colonization will be discussed, including a plausible role for the human microbiome in maintaining a healthy state. With this as a starting point, we will discuss possible disruptive factors contributing to dysbiosis, which is likely to be a key trigger for pathobionts in the development and pathophysiology of respiratory diseases. Finally, from this renewed perspective, we will reflect on current and potential new approaches for treatment in the future.
Collapse
Affiliation(s)
- A P Hakansson
- Division of Experimental Infection Medicine, Department of Translational Medicine, Lund University , Lund , Sweden ; Department of Microbiology, University of Alabama at Birmingham , Birmingham, Alabama ; and Center for Inflammation Research, Queens Medical Research Institute, University of Edinburgh , Edinburgh , United Kingdom
| | - C J Orihuela
- Division of Experimental Infection Medicine, Department of Translational Medicine, Lund University , Lund , Sweden ; Department of Microbiology, University of Alabama at Birmingham , Birmingham, Alabama ; and Center for Inflammation Research, Queens Medical Research Institute, University of Edinburgh , Edinburgh , United Kingdom
| | - D Bogaert
- Division of Experimental Infection Medicine, Department of Translational Medicine, Lund University , Lund , Sweden ; Department of Microbiology, University of Alabama at Birmingham , Birmingham, Alabama ; and Center for Inflammation Research, Queens Medical Research Institute, University of Edinburgh , Edinburgh , United Kingdom
| |
Collapse
|
85
|
Perez AC, Johnson A, Chen Z, Wilding GE, Malkowski MG, Murphy TF. Mapping Protective Regions on a Three-Dimensional Model of the Moraxella catarrhalis Vaccine Antigen Oligopeptide Permease A. Infect Immun 2018; 86:e00652-17. [PMID: 29203544 PMCID: PMC5820933 DOI: 10.1128/iai.00652-17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 11/25/2017] [Indexed: 11/20/2022] Open
Abstract
A vaccine against Moraxella catarrhalis would reduce tremendous morbidity, mortality, and financial burden by preventing otitis media in children and exacerbations of chronic obstructive pulmonary disease (COPD) in adults. Oligopeptide permease A (OppA) is a candidate vaccine antigen that is (i) a nutritional virulence factor expressed on the bacterial cell surface during infection, (ii) widely conserved among strains, (iii) highly immunogenic, and (iv) a protective antigen based on its capacity to induce protective responses in immunized animals. In the present study, we show that the antibodies to OppA following vaccination mediate accelerated clearance in animals after pulmonary challenge. To identify regions of OppA that bind protective antibodies, truncated constructs of OppA were engineered and studied to map regions of OppA with surface-accessible epitopes that bind high-avidity antibodies following vaccination. Protective epitopes were located in the N and C termini of the protein. Immunization of mice with constructs corresponding to these regions (T5 and T8) induced protective responses. Studies of overlapping peptide libraries of constructs T5 and T8 with OppA immune serum identified two discrete regions on each construct. These potentially protective regions were mapped on a three-dimensional computational model of OppA, where regions with solvent-accessible amino acids were identified as three potentially protective epitopes. In all, these studies revealed two regions with three specific epitopes in OppA that induce potentially protective antibody responses following vaccination. Detection of antibodies to these regions could serve to guide vaccine formulation and as a diagnostic tool for monitoring development of protective responses during clinical trials.
Collapse
Affiliation(s)
- Antonia C Perez
- Clinical and Translational Research Center, University at Buffalo, the State University of New York, Buffalo, New York, USA
- Division of Infectious Diseases, Department of Medicine, University at Buffalo, the State University of New York, Buffalo, New York, USA
| | - Antoinette Johnson
- Clinical and Translational Research Center, University at Buffalo, the State University of New York, Buffalo, New York, USA
- Division of Infectious Diseases, Department of Medicine, University at Buffalo, the State University of New York, Buffalo, New York, USA
| | - Ziqiang Chen
- Department of Biostatistics, University at Buffalo, the State University of New York, Buffalo, New York, USA
| | - Gregory E Wilding
- Department of Biostatistics, University at Buffalo, the State University of New York, Buffalo, New York, USA
| | - Michael G Malkowski
- Department of Structural Biology, University at Buffalo, the State University of New York, Buffalo, New York, USA
- Hauptman Woodward Medical Research Institute, Buffalo, New York, USA
| | - Timothy F Murphy
- Clinical and Translational Research Center, University at Buffalo, the State University of New York, Buffalo, New York, USA
- Division of Infectious Diseases, Department of Medicine, University at Buffalo, the State University of New York, Buffalo, New York, USA
- Department of Microbiology, University at Buffalo, the State University of New York, Buffalo, New York, USA
| |
Collapse
|
86
|
Reiss-Mandel A, Rubin C, Zayoud M, Rahav G, Regev-Yochay G. Staphylococcus aureus Colonization Induces Strain-Specific Suppression of Interleukin-17. Infect Immun 2018; 86:e00834-17. [PMID: 29311230 PMCID: PMC5820966 DOI: 10.1128/iai.00834-17] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 12/14/2017] [Indexed: 01/02/2023] Open
Abstract
Staphylococcus aureus is a pathogen that causes significant morbidity and mortality. Nasal carriage is a major source of transmission and of endogenous infection. Persistent carriage is detected in ∼30% of healthy individuals. While Th17 cells have been shown to play a role in S. aureus infection and clearance, the immune response to carriage is not well understood. Here, we evaluate the Th17 response and its potential inhibitors during S. aureus carriage. We recruited 25 volunteers, of whom 11 were persistent carriers. Volunteers' peripheral blood mononuclear cells (PBMCs) were stimulated with either their endogenous strain (a strain isolated from that carrier) or exogenous ones (strains not carried by that volunteer). Changes in Th17 cell frequency and numbers, interleukin-17 (IL-17) mRNA expression, and IL-17 protein abundance were measured by fluorescence-activated cell sorting, real-time PCR, and enzyme-linked immunosorbent assay. Similarly, responses of IL-17 suppressors (regulatory T cells [FOXP3], IL-10, IL-27, and IL-19) were measured. Th17 and IL-17 levels in response to stimulation with endogenous strains were significantly lower than those in response to stimulation with exogenous ones. Of the suppressive cytokines tested, only IL-19 exhibited a stronger response to endogenous than to exogenous strains. Addition of recombinant IL-19 to exogenous-strain-stimulated PBMCs caused decreased IL-17 expression, whereas addition of IL-19 antibodies to endogenous-strain-stimulated cells resulted in an increased IL-17 response. Together, our results suggest that S. aureus carriage induced a tolerogenic response to a carried strain that could be reproduced through the addition of recombinant IL-19 or prevented by the addition of IL-19 antibodies. This differential immune response may play a role in the determination of S. aureus carriage patterns.
Collapse
Affiliation(s)
- Aylana Reiss-Mandel
- Infectious Disease Unit, Sheba Medical Center, Ramat Gan, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Carmit Rubin
- Infectious Disease Unit, Sheba Medical Center, Ramat Gan, Israel
| | - Morad Zayoud
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sheba Cancer Research Center, Sheba Medical Center, Ramat Gan, Israel
| | - Galia Rahav
- Infectious Disease Unit, Sheba Medical Center, Ramat Gan, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gili Regev-Yochay
- Infectious Disease Unit, Sheba Medical Center, Ramat Gan, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
87
|
Moens L, Hermand P, Wellens T, Wuyts G, Derua R, Waelkens E, Ysebaert C, Godfroid F, Bossuyt X. Identification of SP1683 as a pneumococcal protein that is protective against nasopharyngeal colonization. Hum Vaccin Immunother 2018; 14:1234-1242. [PMID: 29400602 DOI: 10.1080/21645515.2018.1430541] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Serotype-independent protein-based pneumococcal vaccines represent attractive alternatives to capsular polysaccharide-based vaccines. The aim of this study was to identify novel immunogenic proteins from Streptococcus pneumoniae that may be used in protein-based pneumococcal vaccine. An immunoproteomics approach and a humanized severe combined immunodeficient mouse model were used to identify S. pneumoniae proteins that are immunogenic for the human immune system. Among the several proteins identified, SP1683 was selected, recombinantly produced, and infection and colonization murine models were used to evaluate the capacity of SP1683 to elicit protective responses, in comparison to known pneumococcal immunogenic proteins (PhtD and detoxified pneumolysin, dPly). Immunisation with SP1683 elicited a weaker antibody response than immunisation with PhtD and did not provide protection in the model of invasive disease. However, similar to PhtD, it was able to significantly reduce colonization in the mouse model of nasopharyngeal carriage. Treatment with anti-IL17A and anti-IL17F antibodies abolished the protection against colonization elicited by SP1683 or PhtD + dPly, which indicated that the protection afforded in this model was Th17-dependent. In conclusion, intranasal immunization with the pneumococcal protein SP1683 conferred IL17-dependent protection against nasopharyngeal carriage in mice, but systemic immunization did not protect against invasive disease. These results do not support the use of SP1683 as an isolated pneumococcal vaccine antigen. Nevertheless, SP1683 could be used as a first line of defence in formulations combining several proteins.
Collapse
Affiliation(s)
- Leen Moens
- a Laboratory of Experimental Laboratory Immunology, Department of Microbiology and Immunology , KU Leuven , Leuven , Belgium
| | | | - Tine Wellens
- a Laboratory of Experimental Laboratory Immunology, Department of Microbiology and Immunology , KU Leuven , Leuven , Belgium
| | - Greet Wuyts
- a Laboratory of Experimental Laboratory Immunology, Department of Microbiology and Immunology , KU Leuven , Leuven , Belgium
| | - Rita Derua
- c Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine , KU Leuven , Leuven , Belgium
| | - Etienne Waelkens
- c Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine , KU Leuven , Leuven , Belgium
| | | | | | - Xavier Bossuyt
- a Laboratory of Experimental Laboratory Immunology, Department of Microbiology and Immunology , KU Leuven , Leuven , Belgium.,d Laboratory Medicine, University Hospitals Leuven , Leuven , Belgium
| |
Collapse
|
88
|
Abstract
Colonization of the human nasopharynx by pneumococcus is extremely common and is both the primary reservoir for transmission and a prerequisite for disease. Current vaccines targeting the polysaccharide capsule effectively prevent colonization, conferring herd protection within vaccinated communities. However, these vaccines cover only a subset of all circulating pneumococcal strains, and serotype replacement has been observed. Given the success of pneumococcal conjugate vaccine (PCV) in preventing colonization in unvaccinated adults within vaccinated communities, reducing nasopharyngeal colonization has become an outcome of interest for novel vaccines. Here, we discuss the immunological mechanisms that control nasopharyngeal colonization, with an emphasis on findings from human studies. Increased understanding of these immunological mechanisms is required to identify correlates of protection against colonization that will facilitate the early testing and design of novel vaccines.
Collapse
Affiliation(s)
- Simon P. Jochems
- Department of Clinicial Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- * E-mail: (SPJ); (DMF)
| | - Jeffrey N. Weiser
- Department of Microbiology, New York University School of Medicine, New York, New York, United States of America
| | - Richard Malley
- Division of Infectious Diseases, Boston Children′s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Daniela M. Ferreira
- Department of Clinicial Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- * E-mail: (SPJ); (DMF)
| |
Collapse
|
89
|
Alnahas S, Hagner S, Raifer H, Kilic A, Gasteiger G, Mutters R, Hellhund A, Prinz I, Pinkenburg O, Visekruna A, Garn H, Steinhoff U. IL-17 and TNF-α Are Key Mediators of Moraxella catarrhalis Triggered Exacerbation of Allergic Airway Inflammation. Front Immunol 2017; 8:1562. [PMID: 29184554 PMCID: PMC5694487 DOI: 10.3389/fimmu.2017.01562] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 10/31/2017] [Indexed: 12/20/2022] Open
Abstract
Alterations of the airway microbiome are often associated with pulmonary diseases. For example, detection of the bacterial pathogen Moraxella catarrhalis in the upper airways is linked with an increased risk to develop or exacerbate asthma. However, the mechanisms by which M. catarrhalis augments allergic airway inflammation (AAI) remain unclear. We here characterized the cellular and soluble mediators of M. catarrhalis triggered excacerbation of AAI in wt and IL-17 deficient as well as in animals treated with TNF-α and IL-6 neutralizing antibodies. We compared the type of inflammatory response in M. catarrhalis infected, house dust mite (HDM)-allergic and animals infected with M. catarrhalis at different time points of HDM sensitization. We found that airway infection of mice with M. catarrhalis triggers a strong inflammatory response with massive neutrophilic infiltrates, high amounts of IL-6 and TNF-α and moderate levels of CD4+ T-cell-derived IFN-γ and IL-17. If bacterial infection occurred during HDM allergen sensitization, the allergic airway response was exacerbated, particularly by the expansion of Th17 cells and increased TNF-α levels. Neutralization of IL-17 or TNF-α but not IL-6 resulted in accelerated clearance of M. catarrhalis and effectively prevented infection-induced exacerbation of AAI. Taken together, our data demonstrate an essential role for TNF-α and IL-17 in infection-triggered exacerbation of AAI.
Collapse
Affiliation(s)
- Safa Alnahas
- Institute of Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| | - Stefanie Hagner
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Member of the German Center for Lung Research, University of Marburg, Marburg, Germany
| | - Hartmann Raifer
- Institute of Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| | - Ayse Kilic
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Member of the German Center for Lung Research, University of Marburg, Marburg, Germany
| | - Georg Gasteiger
- Institute of Medical Microbiology and Hygiene, FZI Research Center for Immunotherapy, University of Mainz Medical Center, Mainz, Germany
| | - Reinier Mutters
- Institute of Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| | - Anne Hellhund
- Institute of Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Olaf Pinkenburg
- Institute of Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| | - Alexander Visekruna
- Institute of Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| | - Holger Garn
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Member of the German Center for Lung Research, University of Marburg, Marburg, Germany
| | - Ulrich Steinhoff
- Institute of Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| |
Collapse
|
90
|
Perez AC, Murphy TF. A Moraxella catarrhalis vaccine to protect against otitis media and exacerbations of COPD: An update on current progress and challenges. Hum Vaccin Immunother 2017; 13:2322-2331. [PMID: 28853985 PMCID: PMC5647992 DOI: 10.1080/21645515.2017.1356951] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/16/2017] [Accepted: 07/12/2017] [Indexed: 01/03/2023] Open
Abstract
Moraxella catarrhalis is a major cause of morbidity and mortality worldwide, especially causing otitis media in young children and exacerbations of chronic obstructive pulmonary disease in adults. This pathogen uses several virulence mechanisms to colonize and survive in its host, including adherence and invasion of host cells, formation of polymicrobial biofilms with other bacterial pathogens, and production of β-lactamase. Given the global impact of otitis media and COPD, an effective vaccine to prevent M. catarrhalis infection would have a huge impact on the quality of life in both patient populations by preventing disease, thus reducing morbidity and health care costs. A number of promising vaccine antigens have been identified for M. catarrhalis. The development of improved animal models of M. catarrhalis disease and identification of a correlate of protection are needed to accelerate vaccine development. This review will discuss the current state of M. catarrhalis vaccine development, and the challenges that must be addressed to succeed.
Collapse
Affiliation(s)
- Antonia C. Perez
- Clinical and Translational Research Center, University at Buffalo, The State University of New York, Buffalo, NY, USA
- Division of Infectious Diseases, Department of Medicine, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Timothy F. Murphy
- Clinical and Translational Research Center, University at Buffalo, The State University of New York, Buffalo, NY, USA
- Division of Infectious Diseases, Department of Medicine, University at Buffalo, The State University of New York, Buffalo, NY, USA
- Department of Microbiology, University at Buffalo, The State University of New York, Buffalo, NY, USA
| |
Collapse
|
91
|
Zafar MA, Wang Y, Hamaguchi S, Weiser JN. Host-to-Host Transmission of Streptococcus pneumoniae Is Driven by Its Inflammatory Toxin, Pneumolysin. Cell Host Microbe 2017; 21:73-83. [PMID: 28081446 DOI: 10.1016/j.chom.2016.12.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/28/2016] [Accepted: 12/06/2016] [Indexed: 01/05/2023]
Abstract
Host-to-host transmission is a critical step for infection. Here we studied transmission of the opportunistic pathogen Streptococcus pneumoniae in an infant mouse model. Transmission from nasally colonized pups required high levels of bacterial shedding in nasal secretions and was temporally correlated with, and dependent upon, the acute inflammatory response. Pneumolysin, a pore-forming cytotoxin and major virulence determinant, was both necessary and sufficient to promote inflammation, which increased shedding and allowed for intralitter transmission. Direct contact between pups was not required for transmission indicating the importance of an environmental reservoir. An additional in vivo effect of pneumolysin was to enhance bacterial survival outside of the host. Our findings provide experimental evidence of a microbial strategy for transit to new hosts and explain why an organism expresses a toxin that damages the host upon which it depends.
Collapse
Affiliation(s)
- M Ammar Zafar
- Department of Microbiology, New York University, New York, NY 10016, USA
| | - Yang Wang
- School of Medicine, Tsinghua University, 100084 Beijing, China; Department of Microbiology, New York University, New York, NY 10016, USA
| | - Shigeto Hamaguchi
- Department of Microbiology, New York University, New York, NY 10016, USA
| | - Jeffrey N Weiser
- Department of Microbiology, New York University, New York, NY 10016, USA.
| |
Collapse
|
92
|
IL-36γ is a crucial proximal component of protective type-1-mediated lung mucosal immunity in Gram-positive and -negative bacterial pneumonia. Mucosal Immunol 2017; 10:1320-1334. [PMID: 28176791 PMCID: PMC5548659 DOI: 10.1038/mi.2016.130] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 12/01/2016] [Indexed: 02/04/2023]
Abstract
Interleukin-36γ (IL-36γ) is a member of novel IL-1-like proinflammatory cytokine family that are highly expressed in epithelial tissues and several myeloid-derived cell types. Little is known about the role of the IL-36 family in mucosal immunity, including lung anti-bacterial responses. We used murine models of IL-36γ deficiency to assess the contribution of IL-36γ in the lung during experimental pneumonia. Induction of IL-36γ was observed in the lung in response to Streptococcus pneumoniae (Sp) infection, and mature IL-36γ protein was secreted primarily in microparticles. IL-36γ-deficient mice challenged with Sp demonstrated increased mortality, decreased lung bacterial clearance and increased bacterial dissemination, in association with reduced local expression of type-1 cytokines, and impaired lung macrophage M1 polarization. IL-36γ directly stimulated type-1 cytokine induction from dendritic cells in vitro in a MyD88-dependent manner. Similar protective effects of IL-36γ were observed in a Gram-negative pneumonia model (Klebsiella pneumoniae). Intrapulmonary delivery of IL-36γ-containing microparticles reconstituted immunity in IL-36γ-/- mice. Enhanced expression of IL-36γ was also observed in plasma and bronchoalveolar lavage fluid of patients with acute respiratory distress syndrome because of pneumonia. These studies indicate that IL-36γ assumes a vital proximal role in the lung innate mucosal immunity during bacterial pneumonia by driving protective type-1 responses and classical macrophage activation.
Collapse
|
93
|
Conjugation of PspA4Pro with Capsular Streptococcus pneumoniae Polysaccharide Serotype 14 Does Not Reduce the Induction of Cross-Reactive Antibodies. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017. [PMID: 28637805 DOI: 10.1128/cvi.00118-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Current pneumococcal vaccines are composed of bacterial polysaccharides as antigens, plain or conjugated to carrier proteins. While efficacious against vaccine serotypes, epidemiologic data show an increasing incidence of infections caused by nonvaccine serotypes of Streptococcus pneumoniae The use of pneumococcal surface protein A (PspA) as a carrier protein in a conjugate vaccine could help prevent serotype replacement by increasing vaccine coverage and reducing selective pressure of S. pneumoniae serotypes. PspA is present in all pneumococcal strains, is highly immunogenic, and is known to induce protective antibodies. Based on its sequence, PspA has been classified into three families and six clades. A PspA fragment derived from family 2, clade 4 (PspA4Pro), was shown to generate antibodies with a broad range of cross-reactivity, across clades and families. Here, PspA4Pro was modified and conjugated to capsular polysaccharide serotype 14 (PS14). We investigated the impact of conjugation on the immune response induced to PspA4Pro and PS14. Mice immunized with the PS14-mPspA4Pro conjugate produced higher titers of anti-PS14 antibodies than the animals that received coadministered antigens. The conjugate induced antibodies with opsonophagocytic activity against PS14-carrying strains, as well as against a panel of strains bearing PspAs from five clades (encompassing families 1 and 2) bearing a non-PS14 serotype. Furthermore, mice immunized with PS14-mPspA4Pro were protected against nasal colonization with a nonrelated S. pneumoniae strain bearing PspA from clade 1, serotype 6B. These results demonstrate that the cross-reactivity mediated by PspA4Pro is retained following conjugation, supporting the use of PspA4 as a carrier protein in order to enhance pneumococcal vaccine coverage and encourage its further investigation as a candidate in future vaccine designs.
Collapse
|
94
|
Jia L, Xie J, Zhao J, Cao D, Liang Y, Hou X, Wang L, Li Z. Mechanisms of Severe Mortality-Associated Bacterial Co-infections Following Influenza Virus Infection. Front Cell Infect Microbiol 2017; 7:338. [PMID: 28824877 PMCID: PMC5540941 DOI: 10.3389/fcimb.2017.00338] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 07/10/2017] [Indexed: 01/15/2023] Open
Abstract
Influenza virus infection remains one of the largest disease burdens on humans. Influenza-associated bacterial co-infections contribute to severe disease and mortality during pandemic and seasonal influenza episodes. The mechanisms of severe morbidity following influenza-bacteria co-infections mainly include failure of an antibacterial immune response and pathogen synergy. Moreover, failure to resume function and tolerance might be one of the main reasons for excessive mortality. In this review, recent advances in the study of mechanisms of severe disease, caused by bacterial co-infections following influenza virus pathogenesis, are summarized. Therefore, understanding the synergy between viruses and bacteria will facilitate the design of novel therapeutic approaches to prevent mortality associated with bacterial co-infections.
Collapse
Affiliation(s)
- Leili Jia
- Institute of Disease Control and Prevention of Chinese People's Liberation ArmyBeijing, China
| | - Jing Xie
- Institute of Disease Control and Prevention of Chinese People's Liberation ArmyBeijing, China
| | - Jiangyun Zhao
- Institute of Disease Control and Prevention of Chinese People's Liberation ArmyBeijing, China
| | - Dekang Cao
- Center for Disease Control and Prevention of Chinese People's Armed Police ForcesBeijing, China
| | - Yuan Liang
- Institute of Disease Control and Prevention of Chinese People's Liberation ArmyBeijing, China
| | - Xuexin Hou
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and PreventionBeijing, China
| | - Ligui Wang
- Institute of Disease Control and Prevention of Chinese People's Liberation ArmyBeijing, China
| | - Zhenjun Li
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and PreventionBeijing, China
| |
Collapse
|
95
|
Ahmed M, Smith DM, Hamouda T, Rangel-Moreno J, Fattom A, Khader SA. A novel nanoemulsion vaccine induces mucosal Interleukin-17 responses and confers protection upon Mycobacterium tuberculosis challenge in mice. Vaccine 2017; 35:4983-4989. [PMID: 28774560 DOI: 10.1016/j.vaccine.2017.07.073] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/13/2017] [Accepted: 07/21/2017] [Indexed: 01/08/2023]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) is contracted via aerosol infection, typically affecting the lungs. Mycobacterium bovis bacillus Calmette-Guerin (BCG) is the only licensed vaccine and has variable efficacy in protecting against pulmonary TB. Additionally, chemotherapy is associated with low compliance contributing to development of multidrug-resistant (MDR) and extensively drug-resistant (XDR) Mtb. Thus, there is an urgent need for the design of more effective vaccines against TB. Experimental vaccines delivered through the mucosal route induce robust T helper type 17 (Th17)/ Interleukin (IL) -17 responses and provide superior protection against Mtb infection. Thus, the development of safe mucosal adjuvants for human use is critical. In this study, we demonstrate that nanoemulsion (NE)-based adjuvants when delivered intranasally along with Mtb specific immunodominant antigens (NE-TB vaccine) induce potent mucosal IL-17T-cell responses. Additionally, the NE-TB vaccine confers significant protection against Mtb infection, and when delivered along with BCG, is associated with decreased disease severity. These findings strongly support the development of a NE-TB vaccine as a novel, safe and effective, first-of-kind IL-17 inducing mucosal vaccine for potential use in humans.
Collapse
Affiliation(s)
- Mushtaq Ahmed
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO 63110, United States
| | | | - Tarek Hamouda
- NanoBio Corporation, Ann Arbor, MI 48105, United States
| | - Javier Rangel-Moreno
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, NY 14624, United States
| | - Ali Fattom
- NanoBio Corporation, Ann Arbor, MI 48105, United States
| | - Shabaana A Khader
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO 63110, United States.
| |
Collapse
|
96
|
Nasopharyngeal polymicrobial colonization during health, viral upper respiratory infection and upper respiratory bacterial infection. J Infect 2017; 75:26-34. [PMID: 28412081 DOI: 10.1016/j.jinf.2017.04.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 04/05/2017] [Accepted: 04/07/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVES We sought to understand how polymicrobial colonization varies during health, viral upper respiratory infection (URI) and acute upper respiratory bacterial infection to understand differences in infection-prone vs. non-prone patients. METHODS Nasopharyngeal (NP) samples were collected from 74 acute otitis media (AOM) infection-prone and 754 non-prone children during 2094 healthy visits, 673 viral URI visits and 631 AOM visits. Three otopathogens Streptococcus pneumoniae (Spn), Nontypeable Haemophilus influenzae (NTHi), and Moraxella catarrhalis (Mcat) were identified by culture. RESULTS NP colonization rates of multiple otopathogens during health were significantly lower than during viral URI, and during URI they were lower than at onset of upper respiratory bacterial infection in both AOM infection-prone and non-prone children. AOM infection-prone children had higher polymicrobial colonization rates than non-prone children during health, viral URI and AOM. Polymicrobial colonization rates of AOM infection-prone children during health were equivalent to that of non-prone children during viral URI, and during viral URI were equivalent to that of non-prone during AOM infection. Spn colonization was positively associated with NTHi and Mcat colonization during health, but negatively during AOM infection. CONCLUSION The infection-prone patients more frequently have multiple potential bacterial pathogens in the NP than the non-prone patients. Polymicrobial interaction in the NP differs during health and at onset of infection.
Collapse
|
97
|
Purified Streptococcus pneumoniae Endopeptidase O (PepO) Enhances Particle Uptake by Macrophages in a Toll-Like Receptor 2- and miR-155-Dependent Manner. Infect Immun 2017; 85:IAI.01012-16. [PMID: 28193634 DOI: 10.1128/iai.01012-16] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 02/03/2017] [Indexed: 12/12/2022] Open
Abstract
Insights into the host-microbial virulence factor interaction, especially the immune signaling mechanisms, could provide novel prevention and treatment options for pneumococcal diseases. Streptococcus pneumoniae endopeptidase O (PepO) is a newly discovered and ubiquitously expressed pneumococcal virulence protein. A PepO-mutant strain showed impaired adherence to and invasion of host cells compared with the isogenic wild-type strain. It is still unknown whether PepO is involved in the host defense response to pneumococcal infection. Here, we demonstrated that PepO could enhance phagocytosis of Streptococcus pneumoniae and Staphylococcus aureus by peritoneal exudate macrophages (PEMs). Further studies showed that PepO stimulation upregulated the expression of microRNA-155 (miR-155) in PEMs in a time- and dose-dependent manner. PepO-induced enhanced phagocytosis was decreased in cells transfected with an inhibitor of miR-155, while it was increased in cells transfected with a mimic of miR-155. We also revealed that PepO-induced upregulation of miR-155 in PEMs was mediated by Toll-like receptor 2 (TLR2)-NF-κB signaling and that the increased expression of miR-155 downregulated expression of SHIP1. Taken together, these results indicate that PepO induces upregulation of miR-155 in PEMs, contributing to enhanced phagocytosis and host defense response to pneumococci and Staphylococcus aureus.
Collapse
|
98
|
Otitis-Prone Children Produce Functional Antibodies to Pneumolysin and Pneumococcal Polysaccharides. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00497-16. [PMID: 28031178 PMCID: PMC5339643 DOI: 10.1128/cvi.00497-16] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 12/14/2016] [Indexed: 12/31/2022]
Abstract
The pneumococcus is a major otitis media (OM) pathogen, but data are conflicting regarding whether otitis-prone children have impaired humoral immunity to pneumococcal antigens. We and others have shown that otitis-prone and healthy children have similar antibody titers to pneumococcal proteins and polysaccharides (vaccine and nonvaccine types); however, the quality of antibodies from otitis-prone children has not been investigated. Antibody function, rather than titer, is considered to be a better correlate of protection from pneumococcal disease. Therefore, we compared the capacities of antibodies from otitis-prone (cases) and healthy (controls) children to neutralize pneumolysin, the pneumococcal toxin currently in development as a vaccine antigen, and to opsonize pneumococcal vaccine and nonvaccine serotypes. A pneumolysin neutralization assay was conducted on cholesterol-depleted complement-inactivated sera from 165 cases and 61 controls. A multiplex opsonophagocytosis assay (MOPA) was conducted on sera from 20 cases and 20 controls. Neutralizing and opsonizing titers were calculated with antigen-specific IgG titers to determine antibody potency for pneumolysin, pneumococcal conjugate vaccine (PCV) polysaccharides, and non-PCV polysaccharides. There was no significant difference in antibody potencies between cases and controls for the antigens tested. Antipneumolysin neutralizing titers increased with the number of episodes of acute OM, but antibody potency did not. Pneumolysin antibody potency was lower in children colonized with pneumococci than in noncarriers, and this was significant for the otitis-prone group (P < 0.05). The production of functional antipneumococcal antibodies in otitis-prone children demonstrates that they respond to the current PCV and are likely to respond to pneumolysin-based vaccines as effectively as healthy children.
Collapse
|
99
|
Ahmed M, Jiao H, Domingo-Gonzalez R, Das S, Griffiths KL, Rangel-Moreno J, Nagarajan UM, Khader SA. Rationalized design of a mucosal vaccine protects against Mycobacterium tuberculosis challenge in mice. J Leukoc Biol 2017; 101:1373-1381. [PMID: 28258153 DOI: 10.1189/jlb.4a0616-270r] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 01/23/2017] [Accepted: 02/15/2017] [Indexed: 12/20/2022] Open
Abstract
Pulmonary tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) is a leading cause of global morbidity and mortality. The only licensed TB vaccine, Mycobacterium bovis bacillus Calmette-Guerin (BCG), has variable efficacy in protecting against pulmonary TB. Thus, the development of more effective TB vaccines is critical to control the TB epidemic. Specifically, vaccines delivered through the mucosal route are known to induce Th17 responses and provide superior protection against Mtb infection. However, already tested Th17-inducing mucosal adjuvants, such as heat-labile enterotoxins and cholera toxins, are not considered safe for use in humans. In the current study, we rationally screened adjuvants for their ability to induce Th17-polarizing cytokines in dendritic cells (DCs) and determined whether they could be used in a protective mucosal TB vaccine. Our new studies show that monophosphoryl lipid A (MPL), when used in combination with chitosan, potently induces Th17-polarizing cytokines in DCs and downstream Th17/Th1 mucosal responses and confers significant protection in mice challenged with a clinical Mtb strain. Additionally, we show that both TLRs and the inflammasome pathways are activated in DCs by MPL-chitosan to mediate induction of Th17-polarizing cytokines. Together, our studies put forward the potential of a new, protective mucosal TB vaccine candidate, which incorporates safe adjuvants already approved for use in humans.
Collapse
Affiliation(s)
- Mushtaq Ahmed
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Hongmei Jiao
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA.,School of Medicine, Yangzhou University, Yangzhou, People's Republic of China
| | - Racquel Domingo-Gonzalez
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Shibali Das
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Kristin L Griffiths
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Javier Rangel-Moreno
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, University of Rochester Medical Center, Rochester, New York, USA; and
| | - Uma M Nagarajan
- Department of Pediatrics and Microbiology/Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Shabaana A Khader
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA;
| |
Collapse
|
100
|
Domínguez-Hüttinger E, Boon NJ, Clarke TB, Tanaka RJ. Mathematical Modeling of Streptococcus pneumoniae Colonization, Invasive Infection and Treatment. Front Physiol 2017; 8:115. [PMID: 28303104 PMCID: PMC5332394 DOI: 10.3389/fphys.2017.00115] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 02/13/2017] [Indexed: 12/26/2022] Open
Abstract
Streptococcus pneumoniae (Sp) is a commensal bacterium that normally resides on the upper airway epithelium without causing infection. However, factors such as co-infection with influenza virus can impair the complex Sp-host interactions and the subsequent development of many life-threatening infectious and inflammatory diseases, including pneumonia, meningitis or even sepsis. With the increased threat of Sp infection due to the emergence of new antibiotic resistant Sp strains, there is an urgent need for better treatment strategies that effectively prevent progression of disease triggered by Sp infection, minimizing the use of antibiotics. The complexity of the host-pathogen interactions has left the full understanding of underlying mechanisms of Sp-triggered pathogenesis as a challenge, despite its critical importance in the identification of effective treatments. To achieve a systems-level and quantitative understanding of the complex and dynamically-changing host-Sp interactions, here we developed a mechanistic mathematical model describing dynamic interplays between Sp, immune cells, and epithelial tissues, where the host-pathogen interactions initiate. The model serves as a mathematical framework that coherently explains various in vitro and in vitro studies, to which the model parameters were fitted. Our model simulations reproduced the robust homeostatic Sp-host interaction, as well as three qualitatively different pathogenic behaviors: immunological scarring, invasive infection and their combination. Parameter sensitivity and bifurcation analyses of the model identified the processes that are responsible for qualitative transitions from healthy to such pathological behaviors. Our model also predicted that the onset of invasive infection occurs within less than 2 days from transient Sp challenges. This prediction provides arguments in favor of the use of vaccinations, since adaptive immune responses cannot be developed de novo in such a short time. We further designed optimal treatment strategies, with minimal strengths and minimal durations of antibiotics, for each of the three pathogenic behaviors distinguished by our model. The proposed mathematical framework will help to design better disease management strategies and new diagnostic markers that can be used to inform the most appropriate patient-specific treatment options.
Collapse
Affiliation(s)
- Elisa Domínguez-Hüttinger
- Department of Bioengineering, Imperial College LondonLondon, UK; Instituto de Ecología, Universidad Nacional Autónoma de MéxicoMexico City, Mexico
| | - Neville J Boon
- Department of Bioengineering, Imperial College London London, UK
| | | | - Reiko J Tanaka
- Department of Bioengineering, Imperial College London London, UK
| |
Collapse
|