1
|
Ercoli G, Selway‐Clarke H, Truijen D, Folkmanaite M, Oulton T, Norris‐Grey C, Nakajima R, Felgner P, Wren BW, Tetteh K, Croucher NJ, Leandro M, Cambridge G, Brown JS. Naturally acquired adaptive immunity to Streptococcus pneumoniae is impaired in rheumatoid arthritis patients. Clin Transl Immunology 2024; 13:e70012. [PMID: 39416767 PMCID: PMC11480415 DOI: 10.1002/cti2.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/10/2024] [Accepted: 10/05/2024] [Indexed: 10/19/2024] Open
Abstract
Objectives Patients with rheumatoid arthritis (RA) have an increased susceptibility to infections, including those caused by Streptococcus pneumoniae. Why RA is associated with increased susceptibility to S. pneumoniae is poorly understood. This study aims to assess the effects of RA and B-cell depletion therapy on naturally acquired antibody responses to 289 S. pneumoniae protein antigens using a novel protein array. Methods IgG responses to S. pneumoniae were characterised in serum from RA patients and disease controls (myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS)) using whole-cell ELISA, a flow cytometry opsonisation assay and an S. pneumoniae protein array. For the RA patients, results were compared before and after B-cell depletion therapy. Results Compared to a well-characterised disease control group of ME/CFS patients, RA patients had reduced antibody responses to multiple S. pneumoniae protein antigens, with significant IgG recognition of approximately half the number of antigens along with reduced median strengths of these responses. Reduction in multiple array antigen-specific responses also correlated with reduced IgG opsonisation of S. pneumoniae. Although B-cell depletion therapy with rituximab did not reduce overall IgG recognition of S. pneumoniae in the RA group, it was associated with marked disruption of pre-existing IgG repertoire to protein antigens in individual patients. Conclusion These data show RA is associated with major disruption of naturally acquired adaptive immunity to S. pneumoniae, which can be assessed rapidly using a protein antigen array and is likely to contribute towards the increased incidence of pneumonia in patients with RA.
Collapse
Affiliation(s)
- Giuseppe Ercoli
- UCL Respiratory, Division of Medicine, Rayne InstituteUniversity College LondonLondonUK
| | - Hugh Selway‐Clarke
- UCL Respiratory, Division of Medicine, Rayne InstituteUniversity College LondonLondonUK
| | - Dena Truijen
- UCL Respiratory, Division of Medicine, Rayne InstituteUniversity College LondonLondonUK
| | - Milda Folkmanaite
- UCL Respiratory, Division of Medicine, Rayne InstituteUniversity College LondonLondonUK
| | - Tate Oulton
- Department of Infection BiologyLondon School of Hygiene & Tropical MedicineLondonUK
| | - Caitlin Norris‐Grey
- Centre for Rheumatology and Bloomsbury Rheumatology Unit, Division of MedicineUniversity College LondonLondonUK
| | - Rie Nakajima
- Vaccine Research and Development Center, Department of Physiology and BiophysicsUniversity of California IrvineIrvineCAUSA
| | - Philip Felgner
- Vaccine Research and Development Center, Department of Physiology and BiophysicsUniversity of California IrvineIrvineCAUSA
| | - Brendan W Wren
- Department of Infection BiologyLondon School of Hygiene & Tropical MedicineLondonUK
| | - Kevin Tetteh
- Department of Infection BiologyLondon School of Hygiene & Tropical MedicineLondonUK
| | - Nicholas J Croucher
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease EpidemiologySchool of Public Health, Imperial College LondonLondonUK
| | - Maria Leandro
- Centre for Rheumatology and Bloomsbury Rheumatology Unit, Division of MedicineUniversity College LondonLondonUK
| | - Geraldine Cambridge
- Centre for Rheumatology and Bloomsbury Rheumatology Unit, Division of MedicineUniversity College LondonLondonUK
| | - Jeremy S Brown
- UCL Respiratory, Division of Medicine, Rayne InstituteUniversity College LondonLondonUK
| |
Collapse
|
2
|
Xiao J, Liu B, Yin Y, Zhang X. Immunization with recombinant Streptococcus pneumoniae PgdA protects mice against lung invasion. Exp Biol Med (Maywood) 2024; 249:10119. [PMID: 39469203 PMCID: PMC11513273 DOI: 10.3389/ebm.2024.10119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
Current pneumococcal vaccines, including the pneumococcal polysaccharide (PPV23) and conjugate (PCV13) vaccines, offer protection against specific serotypes but pose risks of serotype replacement that can alter the composition of the nasopharyngeal microbiota. To address this challenge, a novel strategy has been proposed to provide effective protection without disrupting the colonization of other bacterial populations. In our study, we found that subcutaneous immunization with recombinant peptidoglycan N-acetylglucosamine deacetylase A (rPgdA) elicited robust humoral and cellular immune responses, significantly reducing the invasion of Streptococcus pneumoniae in the lungs without affecting nasopharyngeal carriage. Furthermore, rPgdA antisera were shown to diminish bacterial invasion of lung epithelial cells in vitro. Notably, sera from patients with invasive pneumococcal infections exhibited higher levels of antibodies against the PgdA protein compared to sera from healthy adults, suggesting that a natural immune response to this protein occurs during infection. These results suggest a promising new target for the development of pneumococcal vaccines.
Collapse
Affiliation(s)
- Jiangming Xiao
- Department of Laboratory Medicine, Chongqing General Hospital, Chongqing University, Chongqing, China
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Bichen Liu
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Yibing Yin
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Xuemei Zhang
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Zhao X, Shi W, Li Z, Zhang W. Linking reproductive tract microbiota to premature ovarian insufficiency: Pathophysiological mechanisms and therapies. J Reprod Immunol 2024; 166:104325. [PMID: 39265315 DOI: 10.1016/j.jri.2024.104325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/06/2024] [Accepted: 09/01/2024] [Indexed: 09/14/2024]
Abstract
Over the past decade, research on the human microbiota has become a hot topic. Among them, the female reproductive tract (FRT) also has a specific microbiota that maintains the body's health and dynamic balance, especially in the reproductive aspect. When the FRT ecosystem is dysregulated, changes in immune and metabolic signals can lead to pathological and physiological changes such as chronic inflammation, epithelial barrier disruption, changes in cell proliferation and apoptosis, and dysregulation of angiogenesis and metabolism, thereby causing disruption of the female endocrine system. Premature ovarian insufficiency (POI), a clinical syndrome of ovarian dysfunction, is primarily influenced by immune, genetic, and environmental factors. New evidence suggests that dysbiosis of the FRT microbiota and/or the presence of specific bacteria may contribute to the occurrence and progression of POI. This influence occurs through both direct and indirect mechanisms, including the regulation of estrogen metabolism. The use of probiotics or microbiota transplantation to regulate the microbiome has also been proven to be beneficial in improving ovarian function and the quality of life in women with premature aging. This article provides an overview of the interrelationships and roles between the FRT microbiome and POI in recent years, to fully understand the risk factors affecting female reproductive health, and to offer insights for the future diagnosis, treatment, and application of the FRT microbiome in POI patients.
Collapse
Affiliation(s)
- Xi Zhao
- The First Affiliated Hospital of Hunan University of traditional Chinese medicine, Changsha, Hunan 410000, PR China.
| | - Wenying Shi
- The First Affiliated Hospital of Hunan University of traditional Chinese medicine, Changsha, Hunan 410000, PR China.
| | - Zhengyu Li
- The First Affiliated Hospital of Hunan University of traditional Chinese medicine, Changsha, Hunan 410000, PR China.
| | - Wei Zhang
- The First Affiliated Hospital of Hunan University of traditional Chinese medicine, Changsha, Hunan 410000, PR China.
| |
Collapse
|
4
|
Ahmad I, Burton R, Nahm M, Ejaz HG, Arshad R, Younis BB, Mirza S. Naturally acquired antibodies against 4 Streptococcus pneumoniae serotypes in Pakistani adults with type 2 diabetes mellitus. PLoS One 2024; 19:e0306921. [PMID: 39121085 PMCID: PMC11315336 DOI: 10.1371/journal.pone.0306921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 06/24/2024] [Indexed: 08/11/2024] Open
Abstract
Immune response elicited during pneumococcal carriage has been shown to protect against subsequent colonization and infection by Streptococcus pneumoniae. The study was designed to measure the baseline serotype-specific anti-capsular IgG concentration and opsonic titers elicited in response to asymptomatic carriage in adults with and without type 2-diabetes. Level of IgG to capsular polysaccharide was measured in a total of 176 samples (124 with type 2 diabetes and 52 without type 2 diabetes) against serotype 1, 19F, 9V, and 18C. From within 176 samples, a nested cohort of 39 samples was selected for measuring the functional capacity of antibodies by measuring opsonic titer to serotypes 19F, 9V, and 18C. Next, we measured levels of IgG to PspA in 90 samples from individuals with and without diabetes (22 non-diabetes and 68 diabetes). Our results demonstrated comparable IgG titers against all serotypes between those with and without type 2-diabetes. Overall, we observed higher opsonic titers in those without diabetes as compared to individuals with diabetes for serotypes 19F and 9V. The opsonic titers for 19F and 9V significantly negatively correlated with HbA1c. For 19F, 41.66% (n = 10) showed opsonic titers ≥ 1:8 in the diabetes group as compared to 66.66% (n = 10) in the non-diabetes group. The percentage was 29.6% (n = 7) vs 66.66% (n = 10) for 9V and 70.83% (n = 17) vs 80% (n = 12) for 18C in diabetes and non-diabetes groups respectively. A comparable anti-PspA IgG (p = 0.409) was observed in those with and without diabetes, indicating that response to protein antigen is likely to remain intact in those with diabetes. In conclusion, we demonstrated comparable IgG titers to both capsular polysaccharide and protein antigens in those with and without diabetes, however, the protective capacity of antibodies differed between the two groups.
Collapse
Affiliation(s)
- Izaz Ahmad
- Department of Life Sciences, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Robert Burton
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Moon Nahm
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Hafiz Gohar Ejaz
- Department of Life Sciences, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Rozina Arshad
- Sakina Institute of Diabetes and Endocrine Research, Shalamar Hospital Lahore, Lahore, Pakistan
| | - Bilal Bin Younis
- Sakina Institute of Diabetes and Endocrine Research, Shalamar Hospital Lahore, Lahore, Pakistan
| | - Shaper Mirza
- Department of Life Sciences, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| |
Collapse
|
5
|
Tembo G, Mayuni M, Kamng'ona R, Chimgoneko L, Chiwala G, Sichone S, Galafa B, Thole F, Mkandawire C, Chirwa AE, Nsomba E, Nkhoma V, Ngoliwa C, Toto N, Makhaza L, Muyaya A, Kudowa E, Henrion MYR, Dula D, Morton B, Chikaonda T, Gordon SB, Jambo KC. Poor association between 13-valent pneumococcal conjugate vaccine-induced serum and mucosal antibody responses with experimental Streptococcus pneumoniae serotype 6B colonisation. Vaccine 2024; 42:2975-2982. [PMID: 38570270 PMCID: PMC11056720 DOI: 10.1016/j.vaccine.2024.03.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/23/2024] [Accepted: 03/21/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Pneumococcal carriage is the primary reservoir for transmissionand a prerequisite for invasive pneumococcal disease. Pneumococcal Conjugate Vaccine 13 (PCV13) showed a 62% efficacy in protection against experimental Streptococcus pneumoniae serotype 6B (Spn6B) carriage in a controlled human infection model (CHIM) of healthy Malawian adults. We, therefore, measured humoral responses to experimental challenge and PCV-13 vaccination and determined the association with protection against pneumococcal carriage. METHODS We vaccinated 204 young, healthy Malawian adults with PCV13 or placebo and nasally inoculated them with Spn6B at least four weeks post-vaccination to establish carriage. We collected peripheral blood and nasal lining fluid at baseline, 4 weeks post-vaccination (7 days pre-inoculation), 2, 7, 14 and > 1 year post-inoculation. We measured the concentration of anti-serotype 6B Capsular Polysaccharide (CPS) Immunoglobulin G (IgG) and IgA antibodies in serum and nasal lining fluid using the World Health Organization (WHO) standardised enzyme-linked immunosorbent assay (ELISA). RESULTS PCV13-vaccinated adults had higher serum IgG and nasal IgG/IgA anti-Spn6B CPS-specific binding antibodies than placebo recipients 4 to 6 weeks post-vaccination, which persisted for at least a year after vaccination. Nasal challenge with Spn6B did not significantly alter serum or nasal anti-CPS IgG binding antibody titers with or without experimental pneumococcal carriage. Pre-challenge titers of PCV13-induced serum IgG and nasal IgG/IgA anti-Spn6B CPS binding antibodies did not significantly differ between those that got experimentally colonised by Spn6B compared to those that did not. CONCLUSION This study demonstrates that despite high PCV13 efficacy against experimental Spn6B carriage in young, healthy Malawian adults, robust vaccine-induced systemic and mucosal anti-Spn6B CPS binding antibodies did not directly relate to protection.
Collapse
Affiliation(s)
- G Tembo
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi.
| | - M Mayuni
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - R Kamng'ona
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - L Chimgoneko
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - G Chiwala
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - S Sichone
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - B Galafa
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - F Thole
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - C Mkandawire
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - A E Chirwa
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - E Nsomba
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - V Nkhoma
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - C Ngoliwa
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - N Toto
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - L Makhaza
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - A Muyaya
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - E Kudowa
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - M Y R Henrion
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi; Liverpool School of Tropical Medicine, Clinical Sciences Department, Pembroke Place, Liverpool, UK
| | - D Dula
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - B Morton
- Liverpool School of Tropical Medicine, Clinical Sciences Department, Pembroke Place, Liverpool, UK
| | - T Chikaonda
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - S B Gordon
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi; Liverpool School of Tropical Medicine, Clinical Sciences Department, Pembroke Place, Liverpool, UK
| | - K C Jambo
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi; Liverpool School of Tropical Medicine, Clinical Sciences Department, Pembroke Place, Liverpool, UK.
| |
Collapse
|
6
|
Etesami NS, Barker KA, Shenoy AT, De Ana CL, Arafa EI, Grifno GN, Matschulat AM, Vannini ME, Pihl RMF, Breen MP, Soucy AM, Goltry WN, Ha CT, Betsuyaku H, Browning JL, Varelas X, Traber KE, Jones MR, Quinton LJ, Maglione PJ, Nia HT, Belkina AC, Mizgerd JP. B cells in the pneumococcus-infected lung are heterogeneous and require CD4 + T cell help including CD40L to become resident memory B cells. Front Immunol 2024; 15:1382638. [PMID: 38715601 PMCID: PMC11074383 DOI: 10.3389/fimmu.2024.1382638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/01/2024] [Indexed: 05/12/2024] Open
Abstract
Recovery from respiratory pneumococcal infections generates lung-localized protection against heterotypic bacteria, mediated by resident memory lymphocytes. Optimal protection in mice requires re-exposure to pneumococcus within days of initial infection. Serial surface marker phenotyping of B cell populations in a model of pneumococcal heterotypic immunity revealed that bacterial re-exposure stimulates the immediate accumulation of dynamic and heterogeneous populations of B cells in the lung, and is essential for the establishment of lung resident memory B (BRM) cells. The B cells in the early wave were activated, proliferating locally, and associated with both CD4+ T cells and CXCL13. Antagonist- and antibody-mediated interventions were implemented during this early timeframe to demonstrate that lymphocyte recirculation, CD4+ cells, and CD40 ligand (CD40L) signaling were all needed for lung BRM cell establishment, whereas CXCL13 signaling was not. While most prominent as aggregates in the loose connective tissue of bronchovascular bundles, morphometry and live lung imaging analyses showed that lung BRM cells were equally numerous as single cells dispersed throughout the alveolar septae. We propose that CD40L signaling from antigen-stimulated CD4+ T cells in the infected lung is critical to establishment of local BRM cells, which subsequently protect the airways and parenchyma against future potential infections.
Collapse
Affiliation(s)
- Neelou S. Etesami
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Kimberly A. Barker
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Anukul T. Shenoy
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Carolina Lyon De Ana
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Emad I. Arafa
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Gabrielle N. Grifno
- Department of Biomedical Engineering, Boston University College of Engineering, Boston, MA, United States
| | - Adeline M. Matschulat
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Michael E. Vannini
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Riley M. F. Pihl
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Michael P. Breen
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Alicia M. Soucy
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Wesley N. Goltry
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Catherine T. Ha
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Hanae Betsuyaku
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Jeffrey L. Browning
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Xaralabos Varelas
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Katrina E. Traber
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Matthew R. Jones
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Lee J. Quinton
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Division of Infectious Diseases and Immunology, University of Massachusetts Chan Medical School, Worcester, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Paul J. Maglione
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Hadi T. Nia
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Biomedical Engineering, Boston University College of Engineering, Boston, MA, United States
| | - Anna C. Belkina
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Flow Cytometry Core Facility, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Joseph P. Mizgerd
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| |
Collapse
|
7
|
Majumder S, Li P, Das S, Nafiz TN, Kumar S, Bai G, Dellario H, Sui H, Guan Z, Curtiss R, Furuya Y, Sun W. A bacterial vesicle-based pneumococcal vaccine against influenza-mediated secondary Streptococcus pneumoniae pulmonary infection. Mucosal Immunol 2024; 17:169-181. [PMID: 38215909 PMCID: PMC11033695 DOI: 10.1016/j.mucimm.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/29/2023] [Accepted: 01/08/2024] [Indexed: 01/14/2024]
Abstract
Streptococcus pneumoniae (Spn) is a common pathogen causing a secondary bacterial infection following influenza, which leads to severe morbidity and mortality during seasonal and pandemic influenza. Therefore, there is an urgent need to develop bacterial vaccines that prevent severe post-influenza bacterial pneumonia. Here, an improved Yersinia pseudotuberculosis strain (designated as YptbS46) possessing an Asd+ plasmid pSMV92 could synthesize high amounts of the Spn pneumococcal surface protein A (PspA) antigen and monophosphoryl lipid A as an adjuvant. The recombinant strain produced outer membrane vesicles (OMVs) enclosing a high amount of PspA protein (designated as OMV-PspA). A prime-boost intramuscular immunization with OMV-PspA induced both memory adaptive and innate immune responses in vaccinated mice, reduced the viral and bacterial burden, and provided complete protection against influenza-mediated secondary Spn infection. Also, the OMV-PspA immunization afforded significant cross-protection against the secondary Spn A66.1 infection and long-term protection against the secondary Spn D39 challenge. Our study implies that an OMV vaccine delivering Spn antigens can be a new promising pneumococcal vaccine candidate.
Collapse
Affiliation(s)
- Saugata Majumder
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Peng Li
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Shreya Das
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Tanvir Noor Nafiz
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Sudeep Kumar
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Guangchun Bai
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| | - Hazel Dellario
- Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Haixin Sui
- Wadsworth Center, New York State Department of Health, Albany, New York, USA
| | - Ziqiang Guan
- Department of Biochemistry, Duke University Medical Center, Durham, North Carolina, USA
| | - Roy Curtiss
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| | - Yoichi Furuya
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA.
| | - Wei Sun
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA.
| |
Collapse
|
8
|
Croucher NJ, Campo JJ, Le TQ, Pablo JV, Hung C, Teng AA, Turner C, Nosten F, Bentley SD, Liang X, Turner P, Goldblatt D. Genomic and panproteomic analysis of the development of infant immune responses to antigenically-diverse pneumococci. Nat Commun 2024; 15:355. [PMID: 38191887 PMCID: PMC10774285 DOI: 10.1038/s41467-023-44584-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 12/19/2023] [Indexed: 01/10/2024] Open
Abstract
Streptococcus pneumoniae (pneumococcus) is a nasopharyngeal commensal and respiratory pathogen. This study characterises the immunoglobulin G (IgG) repertoire recognising pneumococci from birth to 24 months old (mo) in a prospectively-sampled cohort of 63 children using a panproteome array. IgG levels are highest at birth, due to transplacental transmission of maternal antibodies. The subsequent emergence of responses to individual antigens exhibit distinct kinetics across the cohort. Stable differences in the strength of individuals' responses, correlating with maternal IgG concentrations, are established by 6 mo. By 12 mo, children develop unique antibody profiles that are boosted by re-exposure. However, some proteins only stimulate substantial responses in adults. Integrating genomic data on nasopharyngeal colonisation demonstrates rare pneumococcal antigens can elicit strong IgG levels post-exposure. Quantifying such responses to the diverse core loci (DCL) proteins is complicated by cross-immunity between variants. In particular, the conserved N terminus of DCL protein zinc metalloprotease B provokes the strongest early IgG responses. DCL proteins' ability to inhibit mucosal immunity likely explains continued pneumococcal carriage despite hosts' polyvalent antibody repertoire. Yet higher IgG levels are associated with reduced incidence, and severity, of pneumonia, demonstrating the importance of the heterogeneity in response strength and kinetics across antigens and individuals.
Collapse
Affiliation(s)
- Nicholas J Croucher
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, W12 0BZ, UK.
| | - Joseph J Campo
- Antigen Discovery Inc, 1 Technology Drive, Irvine, CA, 92618, USA
| | - Timothy Q Le
- Antigen Discovery Inc, 1 Technology Drive, Irvine, CA, 92618, USA
| | - Jozelyn V Pablo
- Antigen Discovery Inc, 1 Technology Drive, Irvine, CA, 92618, USA
| | - Christopher Hung
- Antigen Discovery Inc, 1 Technology Drive, Irvine, CA, 92618, USA
| | - Andy A Teng
- Antigen Discovery Inc, 1 Technology Drive, Irvine, CA, 92618, USA
| | - Claudia Turner
- Cambodia Oxford Medical Research Unit, Angkor Hospital for Children, Siem Reap, 9V54+8FQ, Cambodia
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - François Nosten
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, 63110, Thailand
| | - Stephen D Bentley
- Parasites & Microbes, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Xiaowu Liang
- Antigen Discovery Inc, 1 Technology Drive, Irvine, CA, 92618, USA
| | - Paul Turner
- Cambodia Oxford Medical Research Unit, Angkor Hospital for Children, Siem Reap, 9V54+8FQ, Cambodia
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - David Goldblatt
- Great Ormond Street Institute of Child Health, University College London, London, WC1N 1EH, UK
| |
Collapse
|
9
|
Navaeiseddighi Z, Tripathi JK, Guo K, Wang Z, Schmit T, Brooks DR, Allen RA, Hur J, Mathur R, Jurivich D, Khan N. IL-17RA promotes pathologic epithelial inflammation in a mouse model of upper respiratory influenza infection. PLoS Pathog 2023; 19:e1011847. [PMID: 38060620 PMCID: PMC10729944 DOI: 10.1371/journal.ppat.1011847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 12/19/2023] [Accepted: 11/20/2023] [Indexed: 12/20/2023] Open
Abstract
The upper respiratory tract (nasopharynx or NP) is the first site of influenza replication, allowing the virus to disseminate to the lower respiratory tract or promoting community transmission. The host response in the NP regulates an intricate balance between viral control and tissue pathology. The hyper-inflammatory responses promote epithelial injury, allowing for increased viral dissemination and susceptibility to secondary bacterial infections. However, the pathologic contributors to influenza upper respiratory tissue pathology are incompletely understood. In this study, we investigated the role of interleukin IL-17 recetor A (IL-17RA) as a modulator of influenza host response and inflammation in the upper respiratory tract. We used a combined experimental approach involving IL-17RA-/- mice and an air-liquid interface (ALI) epithelial culture model to investigate the role of IL-17 response in epithelial inflammation, barrier function, and tissue pathology. Our data show that IL-17RA-/- mice exhibited significantly reduced neutrophilia, epithelial injury, and viral load. The reduced NP inflammation and epithelial injury in IL-17RA-/- mice correlated with increased resistance against co-infection by Streptococcus pneumoniae (Spn). IL-17A treatment, while potentiating the apoptosis of IAV-infected epithelial cells, caused bystander cell death and disrupted the barrier function in ALI epithelial model, supporting the in vivo findings.
Collapse
Affiliation(s)
- Zahrasadat Navaeiseddighi
- Dept of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, United States of America
| | - Jitendra Kumar Tripathi
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Kai Guo
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Zhihan Wang
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Taylor Schmit
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Delano R. Brooks
- Dept of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, United States of America
| | - Reese A. Allen
- Dept of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, United States of America
| | - Junguk Hur
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Ramkumar Mathur
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Donald Jurivich
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Nadeem Khan
- Dept of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, United States of America
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| |
Collapse
|
10
|
Zhou S, Lv M, Bai S, Chen W, Zhao W, Wang J, Zhang A, Li J, Xie H, Gao Y, Li D, Wu J. Baseline Pneumococcal IgG Levels and Response to 23-Valent Pneumococcal Polysaccharide Vaccine among Adults from Beijing, China. Vaccines (Basel) 2023; 11:1780. [PMID: 38140184 PMCID: PMC10748153 DOI: 10.3390/vaccines11121780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/19/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
PURPOSE To investigate the baseline levels of serotype-specific IgG antibodies to Streptococcus pneumoniae (S. pneumoniae) and assess their impact on the assessment of vaccine immunogenicity. METHODS We used a subset of serum samples from a randomized controlled trial. The blood of 584 healthy participants was collected on day 0 before and day 28 after the 23-valent pneumococcal polysaccharide vaccine (PPSV23) vaccination. Serotype-specific IgG against PPSV23-covered serotypes were measured by the World Health Organization (WHO) reference enzyme-linked immunosorbent assay (ELISA). Vaccine immunogenicity was compared using conversion rates (proportion of participants with IgG levels following immunization that are 2-fold greater than the baseline) and geometric mean fold rises (GMFRs) between the two groups, which were grouped according to pre-vaccination (baseline) IgG antibody levels. RESULTS Our data showed that over half of individuals have baseline IgG levels for 15 out of 23 serotypes above 1.3 µg/mL, and geometric mean concentrations (GMCs) were generally higher in the elderly group and the female group; significant differences were found in 15 serotypes for vaccine immunogenicity based on the seroconversion rate or GMFRs between individuals with baseline IgG ≥ 1.3 µg/mL and individuals with baseline IgG < 1.3 µg/mL. The seroconversion rate decreased with the increase of baseline IgG levels according to a linear regression model. CONCLUSIONS The assessment of vaccine immunogenicity could be impacted by the fact that many adults had high baseline antibody levels. This study is registered in the Chinese Clinical Trial Registry, number NCT05298800.
Collapse
Affiliation(s)
- Shanshan Zhou
- Beijing Center for Disease Prevention and Control, No. 16, Hepingli Middle Street, Dongcheng District, Beijing 100013, China
| | - Min Lv
- Beijing Center for Disease Prevention and Control, No. 16, Hepingli Middle Street, Dongcheng District, Beijing 100013, China
| | - Shuang Bai
- Beijing Center for Disease Prevention and Control, No. 16, Hepingli Middle Street, Dongcheng District, Beijing 100013, China
| | - Weixin Chen
- Beijing Center for Disease Prevention and Control, No. 16, Hepingli Middle Street, Dongcheng District, Beijing 100013, China
| | - Wei Zhao
- Beijing Center for Disease Prevention and Control, No. 16, Hepingli Middle Street, Dongcheng District, Beijing 100013, China
| | - Jian Wang
- Beijing Center for Disease Prevention and Control, No. 16, Hepingli Middle Street, Dongcheng District, Beijing 100013, China
| | - Ao Zhang
- Beijing Center for Disease Prevention and Control, No. 16, Hepingli Middle Street, Dongcheng District, Beijing 100013, China
| | - Jing Li
- Beijing Center for Disease Prevention and Control, No. 16, Hepingli Middle Street, Dongcheng District, Beijing 100013, China
| | - Hui Xie
- Beijing Center for Disease Prevention and Control, No. 16, Hepingli Middle Street, Dongcheng District, Beijing 100013, China
| | - Yanqing Gao
- Daxing District Center for Disease Control and Prevention of Beijing, Beijing 102600, China
| | - Dongmei Li
- Daxing District Center for Disease Control and Prevention of Beijing, Beijing 102600, China
| | - Jiang Wu
- Beijing Center for Disease Prevention and Control, No. 16, Hepingli Middle Street, Dongcheng District, Beijing 100013, China
| |
Collapse
|
11
|
Gallagher KE, Adetifa IMO, Mburu C, Bottomley C, Akech D, Karani A, Pearce E, Wang Y, Kagucia EW, Goldblatt D, Hammitt LL, Scott JAG. Population immunity to pneumococcal serotypes in Kilifi, Kenya, before and 6 years after the introduction of PCV10 with a catch-up campaign: an observational study of cross-sectional serosurveys. THE LANCET. INFECTIOUS DISEASES 2023; 23:1291-1301. [PMID: 37429307 PMCID: PMC7616650 DOI: 10.1016/s1473-3099(23)00206-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/13/2023] [Accepted: 03/27/2023] [Indexed: 07/12/2023]
Abstract
BACKGROUND In Kilifi (Kenya), a pneumococcal conjugate vaccine (PCV10) was introduced in 2011 in infants (aged <1 year, 3 + 0 schedule) with a catch-up campaign in children aged 1-4 years. We aimed to measure the effect of PCV10 on population immunity. METHODS In this observational study, repeated cross-sectional serosurveys were conducted in independent random samples of 500 children younger than 15 years every 2 years between 2009 and 2017. During these surveys, blood samples were collected by venesection. Concentrations of anti-capsular IgGs against vaccine serotypes (VTs) 1, 4, 5, 6B, 7F, 9V, 14, 18C, 19F, and 23F, and against serotypes 6A and 19A, were assayed by ELISA. We plotted the geometric mean concentrations (GMCs) by birth year to visualise age-specific antibody profiles. In infants, IgG concentrations of 0·35 μg/mL or higher were considered protective. FINDINGS Of 3673 volunteers approached, 2152 submitted samples for analysis across the five surveys. Vaccine introduction resulted in an increase in the proportion of young children with protective IgG concentrations, compared with before vaccine introduction (from 0-33% of infants with VT-specific levels over the correlate of protection in 2009, to 60-94% of infants in 2011). However, among those vaccinated in infancy, GMCs of all ten VTs had waned rapidly by the age of 1, but rose again later in childhood. GMCs among children aged 10-14 years were consistently high over time (eg, the range of GMCs across survey rounds were between 0·45 μg/mL and 1·00 μg/mL for VT 23F and between 2·00 μg/mL and 3·11 μg/mL for VT 19F). INTERPRETATION PCV10 in a 3 + 0 schedule elicited protective IgG levels during infancy, when disease risk is high. The high antibody levels in children aged 10-14 years might indicate continued exposure to vaccine serotypes due to residual carriage or to memory responses to cross-reactive antigens. Despite rapid waning of IgG after vaccination, disease incidence among young children in this setting remains low, suggesting that lower thresholds of antibody, or other markers of immunity (eg, memory B cells), may be needed to assess population protection among children who have aged past infancy. FUNDING Gavi, the Vaccine Alliance; Wellcome Trust.
Collapse
Affiliation(s)
- Katherine E Gallagher
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya; Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK.
| | - Ifedayo M O Adetifa
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya; Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | | | - Christian Bottomley
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Donald Akech
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Angela Karani
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Emma Pearce
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Yanyun Wang
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | | | - David Goldblatt
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Laura L Hammitt
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya; Department of International Health, International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - J Anthony G Scott
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya; Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
12
|
Govindan V, Ganaie FA, Ramakrishnan SM, Ravindran S, Mavuppadi AM, Ravikumar KL. Estimation of baseline IgG antibody levels to 23 pneumococcal vaccine-type capsular polysaccharides in healthy vaccine naïve Indian adults. Vaccine 2023:S0264-410X(23)00497-8. [PMID: 37173269 DOI: 10.1016/j.vaccine.2023.04.074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 04/26/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023]
Abstract
Since immunological responses to pneumococcal vaccines are assessed by a fold-increase in antibody levels relative to pre-immunization levels, it is therefore critical to determine baseline antibody levels to establish putative threshold as a measure of normal response. Herein, for the first time, we measured baseline IgG antibody levels in 108 healthy unvaccinated Indian adults using WHO-recommended ELISA. Median baseline IgG concentration ranged between 0.54 µg/mL to 12.35 µg/mL. Highest levels of baseline capsule polysaccharide (cPS)-specific IgG were found against types 14, 19A, and 33F. Whereas, lowest baseline IgG levels were observed against types 3, 4, and 5. Overall, ∼79 % of study population had median baseline IgG levels ≥1.3 µg/mL against 74 % of cPS's. Substantial baseline antibody levels in unvaccinated adults were observed. The study would be critical in bridging gaps in baseline immunogenicity data and may offer a valuable foundation for evaluating immune response of Indian adults to pneumococcal vaccination.
Collapse
Affiliation(s)
- Vandana Govindan
- Central Research Laboratory, Kempegowda Institute of Medical Sciences, Bangalore, India
| | - Feroze A Ganaie
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care, University of Alabama at Birmingham, AL, USA
| | - Shincy M Ramakrishnan
- Central Research Laboratory, Kempegowda Institute of Medical Sciences, Bangalore, India
| | - Shilpa Ravindran
- Central Research Laboratory, Kempegowda Institute of Medical Sciences, Bangalore, India
| | - Akhila M Mavuppadi
- Central Research Laboratory, Kempegowda Institute of Medical Sciences, Bangalore, India
| | - K L Ravikumar
- Central Research Laboratory, Kempegowda Institute of Medical Sciences, Bangalore, India.
| |
Collapse
|
13
|
Ciacchi L, van de Garde MDB, Ladell K, Farenc C, Poelen MCM, Miners KL, Llerena C, Reid HH, Petersen J, Price DA, Rossjohn J, van Els CACM. CD4 + T cell-mediated recognition of a conserved cholesterol-dependent cytolysin epitope generates broad antibacterial immunity. Immunity 2023; 56:1082-1097.e6. [PMID: 37100059 DOI: 10.1016/j.immuni.2023.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/31/2022] [Accepted: 03/30/2023] [Indexed: 04/28/2023]
Abstract
CD4+ T cell-mediated immunity against Streptococcus pneumoniae (pneumococcus) can protect against recurrent bacterial colonization and invasive pneumococcal diseases (IPDs). Although such immune responses are common, the pertinent antigens have remained elusive. We identified an immunodominant CD4+ T cell epitope derived from pneumolysin (Ply), a member of the bacterial cholesterol-dependent cytolysins (CDCs). This epitope was broadly immunogenic as a consequence of presentation by the pervasive human leukocyte antigen (HLA) allotypes DPB1∗02 and DPB1∗04 and recognition via architecturally diverse T cell receptors (TCRs). Moreover, the immunogenicity of Ply427-444 was underpinned by core residues in the conserved undecapeptide region (ECTGLAWEWWR), enabling cross-recognition of heterologous bacterial pathogens expressing CDCs. Molecular studies further showed that HLA-DP4-Ply427-441 was engaged similarly by private and public TCRs. Collectively, these findings reveal the mechanistic determinants of near-global immune focusing on a trans-phyla bacterial epitope, which could inform ancillary strategies to combat various life-threatening infectious diseases, including IPDs.
Collapse
Affiliation(s)
- Lisa Ciacchi
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Martijn D B van de Garde
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Utrecht 3721MA, the Netherlands
| | - Kristin Ladell
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff CF14 4XN, UK
| | - Carine Farenc
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Martien C M Poelen
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Utrecht 3721MA, the Netherlands
| | - Kelly L Miners
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff CF14 4XN, UK
| | - Carmen Llerena
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Hugh H Reid
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Jan Petersen
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff CF14 4XN, UK; Systems Immunity Research Institute, Cardiff University School of Medicine, University Hospital of Wales, Cardiff CF14 4XN, UK.
| | - Jamie Rossjohn
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff CF14 4XN, UK; Systems Immunity Research Institute, Cardiff University School of Medicine, University Hospital of Wales, Cardiff CF14 4XN, UK.
| | - Cécile A C M van Els
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Utrecht 3721MA, the Netherlands; Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584CL, the Netherlands.
| |
Collapse
|
14
|
Wu Y, Mascaro S, Bhuiyan M, Fathima P, Mace AO, Nicol MP, Richmond PC, Kirkham LA, Dymock M, Foley DA, McLeod C, Borland ML, Martin A, Williams PCM, Marsh JA, Snelling TL, Blyth CC. Predicting the causative pathogen among children with pneumonia using a causal Bayesian network. PLoS Comput Biol 2023; 19:e1010967. [PMID: 36913404 PMCID: PMC10035934 DOI: 10.1371/journal.pcbi.1010967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 03/23/2023] [Accepted: 02/22/2023] [Indexed: 03/14/2023] Open
Abstract
BACKGROUND Pneumonia remains a leading cause of hospitalization and death among young children worldwide, and the diagnostic challenge of differentiating bacterial from non-bacterial pneumonia is the main driver of antibiotic use for treating pneumonia in children. Causal Bayesian networks (BNs) serve as powerful tools for this problem as they provide clear maps of probabilistic relationships between variables and produce results in an explainable way by incorporating both domain expert knowledge and numerical data. METHODS We used domain expert knowledge and data in combination and iteratively, to construct, parameterise and validate a causal BN to predict causative pathogens for childhood pneumonia. Expert knowledge elicitation occurred through a series of group workshops, surveys and one-on-one meetings involving 6-8 experts from diverse domain areas. The model performance was evaluated based on both quantitative metrics and qualitative expert validation. Sensitivity analyses were conducted to investigate how the target output is influenced by varying key assumptions of a particularly high degree of uncertainty around data or domain expert knowledge. RESULTS Designed to apply to a cohort of children with X-ray confirmed pneumonia who presented to a tertiary paediatric hospital in Australia, the resulting BN offers explainable and quantitative predictions on a range of variables of interest, including the diagnosis of bacterial pneumonia, detection of respiratory pathogens in the nasopharynx, and the clinical phenotype of a pneumonia episode. Satisfactory numeric performance has been achieved including an area under the receiver operating characteristic curve of 0.8 in predicting clinically-confirmed bacterial pneumonia with sensitivity 88% and specificity 66% given certain input scenarios (i.e., information that is available and entered into the model) and trade-off preferences (i.e., relative weightings of the consequences of false positive versus false negative predictions). We specifically highlight that a desirable model output threshold for practical use is very dependent upon different input scenarios and trade-off preferences. Three commonly encountered scenarios were presented to demonstrate the potential usefulness of the BN outputs in various clinical pictures. CONCLUSIONS To our knowledge, this is the first causal model developed to help determine the causative pathogen for paediatric pneumonia. We have shown how the method works and how it would help decision making on the use of antibiotics, providing insight into how computational model predictions may be translated to actionable decisions in practice. We discussed key next steps including external validation, adaptation and implementation. Our model framework and the methodological approach can be adapted beyond our context to broad respiratory infections and geographical and healthcare settings.
Collapse
Affiliation(s)
- Yue Wu
- Sydney School of Public Health, University of Sydney, Camperdown, New South Wales, Australia
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Steven Mascaro
- Bayesian Intelligence Pty Ltd, Upwey, Victoria, Australia
- Faculty of Information Technology, Monash University, Clayton, Victoria, Australia
| | - Mejbah Bhuiyan
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Parveen Fathima
- Sydney School of Public Health, University of Sydney, Camperdown, New South Wales, Australia
| | - Ariel O Mace
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
- Department of General Paediaitrics, Perth Children's Hospital, Nedlands, Western Australia, Australia
- Department of Paediatrics, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Mark P Nicol
- School of Biomedical Sciences, University of Western Australia, Crawley, Western Australia, Australia
| | - Peter C Richmond
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
- Department of General Paediaitrics, Perth Children's Hospital, Nedlands, Western Australia, Australia
- School of Medicine, University of Western Australia, Crawley, Western Australia, Australia
| | - Lea-Ann Kirkham
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Michael Dymock
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - David A Foley
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
- Microbiology, PathWest Laboratory Medicine QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Charlie McLeod
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
- Infectious Diseases Department, Perth Children's Hospital, Nedlands, Western Australia, Australia
| | - Meredith L Borland
- School of Medicine, University of Western Australia, Crawley, Western Australia, Australia
- Emergency Department, Perth Children's Hospital, Nedlands, Western Australia, Australia
| | - Andrew Martin
- Department of General Paediaitrics, Perth Children's Hospital, Nedlands, Western Australia, Australia
| | - Phoebe C M Williams
- Sydney School of Public Health, University of Sydney, Camperdown, New South Wales, Australia
- Sydney Children's Hospitals Network, New South Wales, Australia
- School of Women's and Children's Health, The University of New South Wales, Kensington, New South Wales, Australia
| | - Julie A Marsh
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Thomas L Snelling
- Sydney School of Public Health, University of Sydney, Camperdown, New South Wales, Australia
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
- Sydney Children's Hospitals Network, New South Wales, Australia
- School of Public Health, Curtin University, Bentley, Western Australia, Australia
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Christopher C Blyth
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
- School of Medicine, University of Western Australia, Crawley, Western Australia, Australia
- Microbiology, PathWest Laboratory Medicine QEII Medical Centre, Nedlands, Western Australia, Australia
- Infectious Diseases Department, Perth Children's Hospital, Nedlands, Western Australia, Australia
| |
Collapse
|
15
|
Bartlett B, Lee S, Ludewick HP, Siew T, Verma S, Waterer G, Corrales-Medina VF, Dwivedi G. A multiple comorbidities mouse lung infection model in ApoE‑deficient mice. Biomed Rep 2023; 18:21. [PMID: 36846615 PMCID: PMC9944256 DOI: 10.3892/br.2023.1603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 12/09/2022] [Indexed: 02/09/2023] Open
Abstract
Acute pneumonia is characterised by a period of intense inflammation. Inflammation is now considered to be a key step in atherosclerosis progression. In addition, pre-existing atherosclerotic inflammation is considered to play a role in pneumonia progression and risk. In the present study, a multiple comorbidities murine model was used to study respiratory and systemic inflammation that results from pneumonia in the setting of atherosclerosis. Firstly, a minimal infectious dose of Streptococcus pneumoniae (TIGR4 strain) to produce clinical pneumonia with a low mortality rate (20%) was established. C57Bl/6 ApoE -/- mice were fed a high-fat diet prior to administering intranasally 105 colony forming units of TIGR4 or phosphate-buffered saline (PBS). At days 2, 7 and 28 post inoculation (PI), the lungs of mice were imaged by magnetic resonance imaging (MRI) and positron emission tomography (PET). Mice were euthanised and investigated for changes in lung morphology and changes in systemic inflammation using ELISA, Luminex assay and real-time PCR. TIGR4-inoculated mice presented with varying degrees of lung infiltrate, pleural effusion and consolidation on MRI at all time points up to 28 days PI. Moreover, PET scans identified significantly higher FDG uptake in the lungs of TIGR4-inoculated mice up to 28 days PI. The majority (90%) TIGR4-inoculated mice developed pneumococcal-specific IgG antibody response at 28 days PI. Consistent with these observations, TIGR4-inoculated mice displayed significantly increased inflammatory gene expression [interleukin (IL)-1β and IL-6] in the lungs and significantly increased levels of circulating inflammatory protein (CCL3) at 7 and 28 days PI respectively. The mouse model developed by the authors presents a discovery tool to understand the link between inflammation related to acute infection such as pneumonia and increased risk of cardiovascular disease observed in humans.
Collapse
Affiliation(s)
- Benjamin Bartlett
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia 6150, Australia
- School of Medicine, The University of Western Australia, Perth, Western Australia 6009, Australia
| | - Silvia Lee
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia 6150, Australia
- Department of Microbiology, Pathwest Laboratory Medicine, Perth, Western Australia 6000, Australia
| | - Herbert P. Ludewick
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia 6150, Australia
- Heart and Lung Research Institute, Harry Perkins Institute of Medical Research, Perth, Western Australia 6150, Australia
| | - Teck Siew
- Department of Nuclear Medicine, Fiona Stanley Hospital, Perth, Western Australia 6150, Australia
- Royal Perth Hospital, Perth, Western Australia 6000, Australia
| | - Shipra Verma
- Department of Nuclear Medicine, Fiona Stanley Hospital, Perth, Western Australia 6150, Australia
- Department of Geriatric Medicine, Fiona Stanley Hospital, Perth, Western Australia 6150, Australia
| | - Grant Waterer
- School of Medicine, The University of Western Australia, Perth, Western Australia 6009, Australia
- Royal Perth Hospital, Perth, Western Australia 6000, Australia
| | - Vicente F. Corrales-Medina
- Department of Medicine, University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Girish Dwivedi
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Perth, Western Australia 6150, Australia
- School of Medicine, The University of Western Australia, Perth, Western Australia 6009, Australia
- Department of Cardiology, Fiona Stanley Hospital, Perth, Western Australia 6150, Australia
| |
Collapse
|
16
|
Shekhar S, Brar NK, Petersen FC. Suppressive effect of therapeutic antibiotic regimen on antipneumococcal Th1/Th17 responses in neonatal mice. Pediatr Res 2023; 93:818-826. [PMID: 35778498 DOI: 10.1038/s41390-022-02115-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 04/21/2022] [Accepted: 05/02/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Antibiotics are commonly used in human neonates, but their impact on neonatal T cell immunity remains poorly understood. The aim of this study was to investigate the impact of the antibiotic piperacillin with the beta-lactamase inhibitor tazobactam on neonatal CD4+ and CD8+ T cell responses to Streptococcus pneumoniae. METHODS Splenic and lung cells were isolated from the neonatal mice receiving piperacillin and tazobactam or saline (sham) and cultured with S. pneumoniae to analyze T cell cytokine production by ELISA and flow cytometry. RESULTS Antibiotic exposure to neonatal mice resulted in reduced numbers of CD4+/CD8+ T cells in the spleen and lungs compared to control mice. Upon in vitro stimulation with S. pneumoniae, splenocytes and lung cells from antibiotic-exposed mice produced lower levels of IFN-γ (Th1)/IL-17A (Th17) and IL-17A cytokines, respectively. Flow cytometric analysis revealed that S. pneumoniae-stimulated splenic CD4+ T cells from antibiotic-exposed mice expressed decreased levels of IFN-γ and IL-17A compared to control mice, whereas lung CD4+ T cells produced lower levels of IL-17A. However, no significant difference was observed for IL-4 (Th2) production. CONCLUSIONS Neonatal mice exposure to piperacillin and tazobactam reduces the number of CD4+ and CD8+ T cells, and suppresses Th1 and Th17, but not Th2, responses to S. pneumoniae. IMPACT Exposure of neonatal mice with a combination of piperacillin and tazobactam reduces CD4+/CD8+ T cells in the spleen and lungs. Antibiotic exposure suppresses neonatal Th1 and Th17, but not Th2, responses to Streptococcus pneumoniae. Our findings may have important implications for developing better therapeutic strategies in the neonatal intensive care unit.
Collapse
Affiliation(s)
| | - Navdeep K Brar
- Institute of Oral Biology, University of Oslo, Oslo, Norway
| | | |
Collapse
|
17
|
Weller S, Sterlin D, Fadeev T, Coignard E, de los Aires AV, Goetz C, Fritzen R, Bahuaud M, Batteux F, Gorochov G, Weill JC, Reynaud CA. T-independent responses to polysaccharides in humans mobilize marginal zone B cells prediversified against gut bacterial antigens. Sci Immunol 2023; 8:eade1413. [PMID: 36706172 PMCID: PMC7614366 DOI: 10.1126/sciimmunol.ade1413] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 01/04/2023] [Indexed: 01/29/2023]
Abstract
Marginal zone (MZ) B cells are one of the main actors of T-independent (TI) responses in mice. To identify the B cell subset(s) involved in such responses in humans, we vaccinated healthy individuals with Pneumovax, a model TI vaccine. By high-throughput repertoire sequencing of plasma cells (PCs) isolated 7 days after vaccination and of different B cell subpopulations before and after vaccination, we show that the PC response mobilizes large clones systematically, including an immunoglobulin M component, whose diversification and amplification predated the pneumococcal vaccination. These clones could be mainly traced back to MZ B cells, together with clonally related IgA+ and, to a lesser extent, IgG+CD27+ B cells. Recombinant monoclonal antibodies isolated from large PC clones recognized a wide array of bacterial species from the gut flora, indicating that TI responses in humans largely mobilize MZ and switched B cells that most likely prediversified during mucosal immune responses against bacterial antigens and acquired pneumococcal cross-reactivity through somatic hypermutation.
Collapse
Affiliation(s)
- Sandra Weller
- Université Paris Cité, INSERM U1151, CNRS UMR-8253, Institut Necker Enfants Malades (INEM), F-75015 Paris, France
| | - Delphine Sterlin
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), F-75013 Paris, France
- Département d’Immunologie, Assistance Publique Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, F-75013 Paris, France
| | - Tatiana Fadeev
- Université Paris Cité, INSERM U1151, CNRS UMR-8253, Institut Necker Enfants Malades (INEM), F-75015 Paris, France
| | - Eva Coignard
- Université Paris Cité, INSERM U1151, CNRS UMR-8253, Institut Necker Enfants Malades (INEM), F-75015 Paris, France
| | - Alba Verge de los Aires
- Université Paris Cité, INSERM U1151, CNRS UMR-8253, Institut Necker Enfants Malades (INEM), F-75015 Paris, France
| | - Clara Goetz
- Université Paris Cité, INSERM U1151, CNRS UMR-8253, Institut Necker Enfants Malades (INEM), F-75015 Paris, France
| | - Rémi Fritzen
- Université Paris Cité, INSERM U1151, CNRS UMR-8253, Institut Necker Enfants Malades (INEM), F-75015 Paris, France
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Mathilde Bahuaud
- Université Paris Cité, INSERM U1016, Institut Cochin, F-75014 Paris, France
- Service d’Immunologie Biologique, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, F-75014 Paris, France
| | - Frederic Batteux
- Université Paris Cité, INSERM U1016, Institut Cochin, F-75014 Paris, France
- Service d’Immunologie Biologique, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, F-75014 Paris, France
| | - Guy Gorochov
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), F-75013 Paris, France
- Département d’Immunologie, Assistance Publique Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, F-75013 Paris, France
| | - Jean-Claude Weill
- Université Paris Cité, INSERM U1151, CNRS UMR-8253, Institut Necker Enfants Malades (INEM), F-75015 Paris, France
| | - Claude-Agnès Reynaud
- Université Paris Cité, INSERM U1151, CNRS UMR-8253, Institut Necker Enfants Malades (INEM), F-75015 Paris, France
| |
Collapse
|
18
|
Comparative meta-analysis of host transcriptional response during Streptococcus pneumoniae carriage or infection. Microb Pathog 2022; 173:105816. [DOI: 10.1016/j.micpath.2022.105816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/16/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022]
|
19
|
Swarthout TD, Henrion MYR, Thindwa D, Meiring JE, Mbewe M, Kalizang'Oma A, Brown C, Msefula J, Moyo B, Mataya AA, Barnaba S, Pearce E, Gordon M, Goldblatt D, French N, Heyderman RS. Waning of antibody levels induced by a 13-valent pneumococcal conjugate vaccine, using a 3 + 0 schedule, within the first year of life among children younger than 5 years in Blantyre, Malawi: an observational, population-level, serosurveillance study. THE LANCET. INFECTIOUS DISEASES 2022; 22:1737-1747. [PMID: 36029796 PMCID: PMC10555849 DOI: 10.1016/s1473-3099(22)00438-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Pneumococcal conjugate vaccines (PCVs) induce serotype-specific IgG antibodies, effectively reducing vaccine-serotype carriage and invasive pneumococcal disease (IPD). IgG production wanes approximately 1 month after vaccination in absence of serotype-specific exposure. With uncertainty surrrounding correlate of protection (CoP) estimates and with persistent vaccine-serotype carriage and vaccine-serotype IPD after PCV13 introduction, we aimed to profile population-level immunogenicity among children younger than 5 years in Blantyre, Malawi. METHODS For this serosurveillance study, we used a random subset of samples from a prospective population-based serosurvey in Blantyre, Malawi, done between Dec 16, 2016, and June 27, 2018. Sample selection was based on age category optimisation among children younger than 5 years, adequate sample volume, and available budget. We measured serotype-specific IgGs against the 13 vaccine serotypes (1, 3, 4, 5, 6A, 6B, 7F, 9V, 14, 18C, 19A, 19F, and 23F) and two non-vaccine serotypes (12F and 33F), as well as IgGs against three pneumococcal proteins (PsaA, NanA, and Ply), using ELISA and a direct-binding electrochemiluminescence-based multiplex assay. We estimated population-level, serotype-specific immunogenicity profiles using a linear spline regression model. Analyses included samples stratified to 20 3-month age strata (eg, age <3 months to 57-59 months). FINDINGS We evaluated 638 plasma samples: 556 primary samples and 82 unique secondary samples (each linked to one primary sample). Immunogenicity profiles revealed a consistent pattern among vaccine serotypes except serotype 3: a vaccine-induced IgG peak followed by waning to a nadir and subsequent increase in titre. For serotype 3, we observed no apparent vaccine-induced increase. Heterogeneity in parameters included age range at post-vaccination nadir (from 11·2 months [19A] to 27·3 months [7F]). The age at peak IgG titre ranged from 2·69 months (5) to 6·64 months (14). Titres dropped below CoPs against IPD among nine vaccine serotypes (1, 3, 4, 5, 6B, 7F, 9V, 18C, and 23F) and below CoPs against carriage for ten vaccine serotypes (1, 4, 5, 6B, 7F, 9V, 14, 18C, 19F, and 23F). Increasing antibody concentrations among older children and seroincident events were consistent with ongoing vaccine-serotype exposure. INTERPRETATION A 3 + 0 PCV13 schedule with high uptake has not led to sustained population-level antibody immunity beyond the first year of life. Indeed, post-vaccine antibody concentrations dropped below putative CoPs for several vaccine serotypes, potentially contributing to persistent vaccine-serotype carriage and residual vaccine-serotype IPD in Malawi and other similar settings. Policy decisions should consider alternative vaccine strategies, including a booster dose, to achieve sustained vaccine-induced antibody titres, and thus control. FUNDING Bill & Melinda Gates Foundation, Wellcome UK, and National Institute for Health and Care Research.
Collapse
Affiliation(s)
- Todd D Swarthout
- National Institute for Health and Care Research Mucosal Pathogens Research Unit, Research Department of Infection, Division of Infection and Immunity, University College London, London, UK; Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi.
| | - Marc Y R Henrion
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi; Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Deus Thindwa
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi; Centre for the Mathematical Modelling of Infectious Diseases, Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - James E Meiring
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Maurice Mbewe
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Akuzike Kalizang'Oma
- National Institute for Health and Care Research Mucosal Pathogens Research Unit, Research Department of Infection, Division of Infection and Immunity, University College London, London, UK; Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Comfort Brown
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Jacquline Msefula
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi; Faculty of Medicine, University of Amsterdam, Amsterdam, Netherlands
| | - Brewster Moyo
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Andrew A Mataya
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Susanne Barnaba
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi; Chancellor College, University of Malawi, Blantyre, Malawi
| | - Emma Pearce
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Melita Gordon
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi; Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - David Goldblatt
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Neil French
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Robert S Heyderman
- National Institute for Health and Care Research Mucosal Pathogens Research Unit, Research Department of Infection, Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
20
|
Nguyen PTN, Le NV, Dinh HMN, Nguyen BQP, Nguyen TVA. Lung penetration and pneumococcal target binding of antibiotics in lower respiratory tract infection. Curr Med Res Opin 2022; 38:2085-2095. [PMID: 36189961 DOI: 10.1080/03007995.2022.2131304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVES To achieve the therapeutic effects, antibiotics must penetrate rapidly into infection sites and bind to targets. This study reviewed updated knowledge on the ability of antibiotics to penetrate into the lung, their physicochemical properties influencing the pulmonary penetration and their ability to bind to targets on pneumococci. METHODS A search strategy was developed using PubMED, Web of Science, and ChEMBL. Data on serum protein binding, drug concentration, target binding ability, drug transporters, lung penetration, physicochemical properties of antibiotics in low respiratory tract infection (LRTI) were collected. RESULTS It was seen that infection site-to-serum concentration ratios of most antibiotics are >1 at different time points except for ceftriaxone, clindamycin and vancomycin. Most agents have proper physicochemical properties that facilitate antibiotic penetration. In antimicrobial-resistant Streptococcus pneumoniae, the binding affinity of antibiotics to targets mostly decreases compared to that in susceptible strains. The data on binding affinity of linezolid, clindamycin and vancomycin were insufficient. The higher drug concentration at the infection sites compared to that in the blood can be associated with inflammation conditions. Little evidence showed the effect of drug transporters on the clinical efficacy of antibiotics against LRTI. CONCLUSIONS Data on antibiotic penetration into the lung in LRTI patients and binding affinity of antibiotics for pneumococcal targets are still limited. Further studies are required to clarify the associations of the lung penetration and target binding ability of antibitotics with therapeutic efficacy to help propose the right antibiotics for LRTI.
Collapse
Affiliation(s)
| | - Nho Van Le
- Danang University of Medical Technology and Pharmacy, Da Nang, Vietnam
| | | | | | - Thi Van Anh Nguyen
- Department of Life Sciences, University of Science and Technology of Hanoi (USTH), Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| |
Collapse
|
21
|
Cui Y, Miao C, Chen W, Shang W, Qi Q, Zhou W, Wang X, Li Y, Yan Z, Jiang Y. Construction and protective efficacy of a novel Streptococcus pneumoniae fusion protein vaccine NanAT1-TufT1-PlyD4. Front Immunol 2022; 13:1043293. [PMID: 36389808 PMCID: PMC9659761 DOI: 10.3389/fimmu.2022.1043293] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/17/2022] [Indexed: 01/19/2024] Open
Abstract
During the past decades, with the implementation of pneumococcal polysaccharide vaccine (PPV) and pneumococcal conjugate vaccines (PCVs), a dramatic reduction in vaccine type diseases and transmissions has occurred. However, it is necessary to develop a less expensive, serotype-independent pneumococcal vaccine due to the emergence of nonvaccine-type pneumococcal diseases and the limited effect of vaccines on colonization. As next-generation vaccines, conserved proteins, such as neuraminidase A (NanA), elongation factor Tu (Tuf), and pneumolysin (Ply), are promising targets against pneumococcal infections. Here, we designed and constructed a novel fusion protein, NanAT1-TufT1-PlyD4, using the structural and functional domains of full-length NanA, Tuf and Ply proteins with suitable linkers based on bioinformatics analysis and molecular cloning technology. Then, we tested whether the protein protected against focal and lethal pneumococcal infections and examined its potential protective mechanisms. The fusion protein NanAT1-TufT1-PlyD4 consists of 627 amino acids, which exhibits a relatively high level of thermostability, high stability, solubility and a high antigenic index without allergenicity. The purified fusion protein was used to subcutaneously immunize C57BL/6 mice, and NanAT1-TufT1-PlyD4 induced a strong and significant humoral immune response. The anti-NanAT1-TufT1-PlyD4 specific IgG antibody assays increased after the first immunization and reached the highest value at the 35th day. The results from in vitro experiments showed that anti-NanAT1-TufT1-PlyD4 antisera could inhibit the adhesion of Streptococcus pneumoniae (S. pneumoniae) to A549 cells. In addition, immunization with NanAT1-TufT1-PlyD4 significantly reduced S. pneumoniae colonization in the lung and decreased the damage to the lung tissues induced by S. pneumoniae infection. After challenge with a lethal dose of serotype 3 (NC_WCSUH32403), a better protection effect was observed with NanAT1-TufT1-PlyD4-immunized mice than with the separate full-length proteins and the adjuvant control; the survival rate was 50%, which met the standard of the marketed vaccine. Moreover, we showed that the humoral immune response and the Th1, Th2 and Th17-cellular immune pathways are involved in the immune protection of NanAT1-TufT1-PlyD4 to the host. Collectively, our results support that the novel fusion protein NanAT1-TufT1-PlyD4 exhibits extensive immune stimulation and is effective against pneumococcal challenges, and these properties are partially attributed to humoral and cellular-mediated immune responses.
Collapse
Affiliation(s)
- Yali Cui
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Laboratory Medicine, Meishan Women and Children’s Hospital, Alliance Hospital of West China Second University Hospital, Sichuan University, Meishan, China
| | - Chenglin Miao
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Wen Chen
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Wenling Shang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Qianqian Qi
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Wei Zhou
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xia Wang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Yingying Li
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Ziyi Yan
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Yongmei Jiang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| |
Collapse
|
22
|
Silva PH, Vázquez Y, Campusano C, Retamal-Díaz A, Lay MK, Muñoz CA, González PA, Kalergis AM, Bueno SM. Non-capsular based immunization approaches to prevent Streptococcus pneumoniae infection. Front Cell Infect Microbiol 2022; 12:949469. [PMID: 36225231 PMCID: PMC9548657 DOI: 10.3389/fcimb.2022.949469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 09/02/2022] [Indexed: 12/02/2022] Open
Abstract
Streptococcus pneumoniae is a Gram-positive bacterium and the leading cause of bacterial pneumonia in children and the elderly worldwide. Currently, two types of licensed vaccines are available to prevent the disease caused by this pathogen: the 23-valent pneumococcal polysaccharide-based vaccine and the 7-, 10, 13, 15 and 20-valent pneumococcal conjugate vaccine. However, these vaccines, composed of the principal capsular polysaccharide of leading serotypes of this bacterium, have some problems, such as high production costs and serotype-dependent effectiveness. These drawbacks have stimulated research initiatives into non-capsular-based vaccines in search of a universal vaccine against S. pneumoniae. In the last decades, several research groups have been developing various new vaccines against this bacterium based on recombinant proteins, live attenuated bacterium, inactivated whole-cell vaccines, and other newer platforms. Here, we review and discuss the status of non-capsular vaccines against S. pneumoniae and the future of these alternatives in a post-pandemic scenario.
Collapse
Affiliation(s)
- Pedro H. Silva
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Yaneisi Vázquez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camilo Campusano
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Angello Retamal-Díaz
- Departamento de Biotecnología, Facultad de Ciencias del Mar y Recursos Biológicos, Universidad de Antofagasta, Antofagasta, Chile
| | - Margarita K. Lay
- Departamento de Biotecnología, Facultad de Ciencias del Mar y Recursos Biológicos, Universidad de Antofagasta, Antofagasta, Chile
| | - Christian A. Muñoz
- Unidad de Microbiología, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- *Correspondence: Susan M. Bueno,
| |
Collapse
|
23
|
Davies LRL, Cizmeci D, Guo W, Luedemann C, Alexander-Parrish R, Grant L, Isturiz R, Theilacker C, Jodar L, Gessner BD, Alter G. Polysaccharide and conjugate vaccines to Streptococcus pneumoniae generate distinct humoral responses. Sci Transl Med 2022; 14:eabm4065. [PMID: 35921476 PMCID: PMC9885968 DOI: 10.1126/scitranslmed.abm4065] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Streptococcus pneumoniae is a major cause of community-acquired pneumonia, bacteremia, and meningitis in older adults worldwide. Two pneumococcal vaccines containing S. pneumoniae capsular polysaccharides are in current use: the polysaccharide vaccine PPSV23 and the glycoconjugate vaccine PCV13. In clinical trials, both vaccines elicit similar opsonophagocytic killing activity. In contrast to polysaccharide vaccines, conjugate vaccines have shown consistent efficacy against nasopharyngeal carriage and noninvasive pneumonia overall and for some prevalent individual serotypes. Given these different clinical profiles, it is crucial to understand the differential immunological responses induced by these two vaccines. Here, we used a high-throughput systems serology approach to profile the biophysical and functional features of serum antibodies induced by PCV13 and PPSV23 at 1 month and 1 year. In comparison with PPSV23, PCV13 induced higher titers across antibody isotypes; more durable antibody responses across immunoglobulin G (IgG), IgA, and IgM isotypes; and increased antigenic breadth. Although titers measured in opsonophagocytic activity (OPA) assays were similar between the two groups, confirming what was observed in clinical studies, serum samples from PCV13 vaccinees could induce additional non-OPA antibody-dependent functions, including monocyte phagocytosis and natural killer cell activation. In a multivariate modeling approach, distinct humoral profiles were demonstrated in each arm. Together, these results demonstrate that the glycoconjugate PCV13 vaccine induces an antigenically broader, more durable, polyfunctional antibody response. These findings may help explain the increased protection against S. pneumoniae colonization and noninvasive pneumonia and the longer duration of protection against invasive pneumococcal disease, mediated by PCV13.
Collapse
Affiliation(s)
- Leela R. L. Davies
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
- Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Deniz Cizmeci
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Wenyue Guo
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | | | | | | | | | | | - Luis Jodar
- Pfizer Vaccines, Collegeville, PA 19426, USA
| | | | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| |
Collapse
|
24
|
Chaguza C, Yang M, Jacques LC, Bentley SD, Kadioglu A. Serotype 1 pneumococcus: epidemiology, genomics, and disease mechanisms. Trends Microbiol 2022; 30:581-592. [PMID: 34949516 PMCID: PMC7613904 DOI: 10.1016/j.tim.2021.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 12/14/2022]
Abstract
Streptococcus pneumoniae (the 'pneumococcus') is a significant cause of morbidity and mortality worldwide, causing life-threatening diseases such as pneumonia, bacteraemia, and meningitis, with an annual death burden of over one million. Discovered over a century ago, pneumococcal serotype 1 (S1) is a significant cause of these life-threatening diseases. Our understanding of the epidemiology and biology of pneumococcal S1 has significantly improved over the past two decades, informing the development of preventative and surveillance strategies. However, many questions remain unanswered. Here, we review the current state of knowledge of pneumococcal S1, with a special emphasis on clinical epidemiology, genomics, and disease mechanisms.
Collapse
Affiliation(s)
- Chrispin Chaguza
- Parasites and Microbes Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK; Darwin College, University of Cambridge, Silver Street, Cambridge, UK; Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, The Ronald Ross Building, West Derby St, Liverpool, UK; NIHR Mucosal Pathogens Research Unit, Division of Infection and Immunity, University College London, London, UK.
| | - Marie Yang
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, The Ronald Ross Building, West Derby St, Liverpool, UK
| | - Laura C Jacques
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, The Ronald Ross Building, West Derby St, Liverpool, UK.
| | - Stephen D Bentley
- Parasites and Microbes Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK; Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, The Ronald Ross Building, West Derby St, Liverpool, UK; Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, UK
| | - Aras Kadioglu
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, The Ronald Ross Building, West Derby St, Liverpool, UK
| |
Collapse
|
25
|
Menghani SV, Cutcliffe MP, Sanchez-Rosario Y, Pok C, Watson A, Neubert MJ, Ochoa K, Wu HJJ, Johnson MDL. N, N-Dimethyldithiocarbamate Elicits Pneumococcal Hypersensitivity to Copper and Macrophage-Mediated Clearance. Infect Immun 2022; 90:e0059721. [PMID: 35311543 PMCID: PMC9022595 DOI: 10.1128/iai.00597-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 02/12/2022] [Indexed: 12/26/2022] Open
Abstract
Streptococcus pneumoniae is a Gram-positive, encapsulated bacterium that is a significant cause of disease burden in pediatric and elderly populations. The rise in unencapsulated disease-causing strains and antimicrobial resistance in S. pneumoniae has increased the need for developing new antimicrobial strategies. Recent work by our laboratory has identified N,N-dimethyldithiocarbamate (DMDC) as a copper-dependent antimicrobial against bacterial, fungal, and parasitic pathogens. As a bactericidal antibiotic against S. pneumoniae, DMDC's ability to work as a copper-dependent antibiotic and its ability to work in vivo warranted further investigation. Here, our group studied the mechanisms of action of DMDC under various medium and excess-metal conditions and investigated DMDC's interactions with the innate immune system in vitro and in vivo. Of note, we found that DMDC plus copper significantly increased the internal copper concentration, hydrogen peroxide stress, nitric oxide stress, and the in vitro macrophage killing efficiency and decreased capsule. Furthermore, we found that in vivo DMDC treatment increased the quantity of innate immune cells in the lung during infection. Taken together, this study provides mechanistic insights regarding DMDC's activity as an antibiotic at the host-pathogen interface.
Collapse
Affiliation(s)
- Sanjay V. Menghani
- Department of Immunobiology, University of Arizona College of Medicine—Tucson, Tucson, Arizona, USA
- Medical Scientist Training M.D.-Ph.D. Program (MSTP), University of Arizona College of Medicine—Tucson, Tucson, Arizona, USA
| | - Madeline P. Cutcliffe
- Department of Immunobiology, University of Arizona College of Medicine—Tucson, Tucson, Arizona, USA
| | - Yamil Sanchez-Rosario
- Department of Immunobiology, University of Arizona College of Medicine—Tucson, Tucson, Arizona, USA
| | - Chansorena Pok
- Department of Immunobiology, University of Arizona College of Medicine—Tucson, Tucson, Arizona, USA
| | - Alison Watson
- Department of Immunobiology, University of Arizona College of Medicine—Tucson, Tucson, Arizona, USA
| | - Miranda J. Neubert
- Department of Immunobiology, University of Arizona College of Medicine—Tucson, Tucson, Arizona, USA
| | - Klariza Ochoa
- Department of Immunobiology, University of Arizona College of Medicine—Tucson, Tucson, Arizona, USA
| | - Hsin-Jung Joyce Wu
- Department of Immunobiology, University of Arizona College of Medicine—Tucson, Tucson, Arizona, USA
- Arizona Arthritis Center, University of Arizona College of Medicine—Tucson, Tucson, Arizona, USA
| | - Michael D. L. Johnson
- Department of Immunobiology, University of Arizona College of Medicine—Tucson, Tucson, Arizona, USA
- Valley Fever Center for Excellence, University of Arizona College of Medicine—Tucson, Tucson, Arizona, USA
- BIO5 Institute, University of Arizona College of Medicine—Tucson, Tucson, Arizona, USA
- Asthma and Airway Disease Research Center, University of Arizona College of Medicine—Tucson, Tucson, Arizona, USA
| |
Collapse
|
26
|
Ren S, Hansbro PM, Srikusalanukul W, Horvat JC, Hunter T, Brown AC, Peel R, Faulkner J, Evans TJ, Li SC, Newby D, Hure A, Abhayaratna WP, Tsimikas S, Gonen A, Witztum JL, Attia J, Hansbro PM, Peel R, Srikusalanukul W, Abhayaratna W, Newby D, Hure A, D'Este C, Tonkin A, Hopper I, Thrift A, Levi C, Sturm J, Durrheim D, Hung J, Briffa T, Chew D, Anderson P, Moon L, McEvoy M, Attia J. Generation of cardio-protective antibodies after pneumococcal polysaccharide vaccine: Early results from a randomised controlled trial. Atherosclerosis 2022; 346:68-74. [PMID: 35290813 DOI: 10.1016/j.atherosclerosis.2022.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 01/31/2022] [Accepted: 02/09/2022] [Indexed: 11/02/2022]
Abstract
BACKGROUND AND AIMS Observational studies have demonstrated that the pneumococcal polysaccharide vaccine (PPV) is associated with reduced risk of cardiovascular events. This may be mediated through IgM antibodies to OxLDL, which have previously been associated with cardioprotective effects. The Australian Study for the Prevention through Immunisation of Cardiovascular Events (AUSPICE) is a double-blind, randomised controlled trial (RCT) of PPV in preventing ischaemic events. Participants received PPV or placebo once at baseline and are being followed-up for incident fatal and non-fatal myocardial infarction or stroke over 6 years. METHODS A subgroup of participants at one centre (Canberra; n = 1,001) were evaluated at 1 month and 2 years post immunisation for changes in surrogate markers of atherosclerosis, as pre-specified secondary outcomes: high-sensitive C-reactive protein (CRP), pulse wave velocity (PWV), and carotid intima-media thickness (CIMT). In addition, 100 participants were randomly selected in each of the intervention and control groups for measurement of anti-pneumococcal antibodies (IgG, IgG2, IgM) as well as anti-OxLDL antibodies (IgG and IgM to CuOxLDL, MDA-LDL, and PC-KLH). RESULTS Concentrations of anti-pneumococcal IgG and IgG2 increased and remained high at 2 years in the PPV group compared to the placebo group, while IgM increased and then declined, but remained detectable, at 2 years. There were statistically significant increases in all anti-OxLDL IgM antibodies at 1 month, which were no longer detectable at 2 years; there was no increase in anti-OxLDL IgG antibodies. There were no significant changes in CRP, PWV or CIMT between the treatment groups at the 2-year follow-up. CONCLUSIONS PPV engenders a long-lasting increase in anti-pneumococcal IgG, and to a lesser extent, IgM titres, as well as a transient increase in anti-OxLDL IgM antibodies. However, there were no detectable changes in surrogate markers of atherosclerosis at the 2-year follow-up. Long-term, prospective follow-up of clinical outcomes is continuing to assess if PPV reduces CVD events.
Collapse
Affiliation(s)
- Shu Ren
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia
| | - Philip M Hansbro
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia; Centenary UTS Centre for Inflammation, Sydney, NSW, Australia
| | - Wichat Srikusalanukul
- Australian National University Medical School, Canberra Hospital, Canberra, ACT, Australia
| | - Jay C Horvat
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Tegan Hunter
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Alexandra C Brown
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Roseanne Peel
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia.
| | - Jack Faulkner
- Hunter Medical Research Institute, Newcastle, NSW, Australia
| | | | - Shu Chuen Li
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
| | - David Newby
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
| | - Alexis Hure
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Walter P Abhayaratna
- Australian National University Medical School, Canberra Hospital, Canberra, ACT, Australia
| | - Sotirios Tsimikas
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Ayelet Gonen
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Joseph L Witztum
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - John Attia
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia; Hunter Medical Research Institute, Newcastle, NSW, Australia; Department of Medicine, John Hunter Hospital, Newcastle, NSW, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Ercoli G, Ramos‐Sevillano E, Pearce E, Ragab S, Goldblatt D, Weckbecker G, Brown JS. Maintained partial protection against Streptococcus pneumoniae despite B-cell depletion in mice vaccinated with a pneumococcal glycoconjugate vaccine. Clin Transl Immunology 2022; 11:e1366. [PMID: 35003749 PMCID: PMC8715227 DOI: 10.1002/cti2.1366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/16/2021] [Accepted: 12/16/2021] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVES Anti-CD20 monoclonal antibody therapy rapidly depletes > 95% of CD20+ B cells from the circulation. B-cell depletion is an effective treatment for autoimmune disease and B-cell malignancies but also increases the risk of respiratory tract infections. This effect on adaptive immunity could be countered by vaccination. We have used mouse models to investigate the effects of B-cell depletion on pneumococcal vaccination, including protection against infection and timing of vaccination in relation to B-cell depletion. METHODS C57BL/6 female mice were B-cell depleted using anti-CD20 antibody and immunized with two doses of Prevnar-13 vaccine either before or after anti-CD20 treatment. B-cell repertoire and Streptococcus pneumoniae-specific IgG levels were measured using whole-cell ELISA and flow cytometry antibody-binding assay. Protection induced by vaccination was assessed by challenging the mice using a S. pneumoniae pneumonia model. RESULTS Antibody responses to S. pneumoniae were largely preserved in mice B-cell depleted after vaccination resulting in full protection against pneumococcal infections. In contrast, mice vaccinated with Prevnar-13 while B cells were depleted (with > 90% reduction in B-cell numbers) had decreased circulating anti-S. pneumoniae IgG and IgM levels (measured using ELISA and flow cytometry antibody binding assays). However, some antibody responses were maintained, and, although vaccine-induced protection against S. pneumoniae infection was impaired, septicaemia was still prevented in 50% of challenged mice. CONCLUSIONS This study showed that although vaccine efficacy during periods of profound B-cell depletion was impaired some protective efficacy was preserved, suggesting that vaccination remains beneficial.
Collapse
Affiliation(s)
- Giuseppe Ercoli
- Centre for Inflammation and Tissue RepairUCL RespiratoryDivision of MedicineUniversity College Medical SchoolRayne InstituteLondonUK
| | - Elisa Ramos‐Sevillano
- Centre for Inflammation and Tissue RepairUCL RespiratoryDivision of MedicineUniversity College Medical SchoolRayne InstituteLondonUK
| | - Emma Pearce
- Department of ImmunobiologyUCL Great Ormond Street Institute of Child HealthNIHR Biomedical Research CentreLondonUK
| | - Sara Ragab
- Department of ImmunobiologyUCL Great Ormond Street Institute of Child HealthNIHR Biomedical Research CentreLondonUK
| | - David Goldblatt
- Department of ImmunobiologyUCL Great Ormond Street Institute of Child HealthNIHR Biomedical Research CentreLondonUK
| | | | - Jeremy S Brown
- Centre for Inflammation and Tissue RepairUCL RespiratoryDivision of MedicineUniversity College Medical SchoolRayne InstituteLondonUK
| |
Collapse
|
28
|
Li M, Bou-Dargham MJ, Yu J, Etwebi Z, Sun H, Chen YH. TIPE polarity proteins are required for mucosal deployment of T lymphocytes and mucosal defense against bacterial infection. MOLECULAR BIOMEDICINE 2021; 2:41. [PMID: 34939151 PMCID: PMC8695405 DOI: 10.1186/s43556-021-00059-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/05/2021] [Indexed: 11/18/2022] Open
Abstract
Mucosal surfaces are continuously exposed to, and challenged by, numerous commensal and pathogenic organisms. To guard against infections, a majority of the thymus-derived T lymphocytes are deployed at the mucosa. Although chemokines are known to be involved in the mucosal lymphocyte deployment, it is not clear whether lymphocytes enter the mucosa through directed migration or enhanced random migration. Here we report that TIPE (tumor necrosis factor-α-induced protein 8 (TNFAIP8)-like) proteins mediate directed migration of T lymphocytes into lung mucosa, and they are crucial for mucosal immune defense against Streptococcus pneumoniae infection. Knockout of both Tnfaip8 and Tipe2, which encode polarity proteins that control the directionality of lymphocyte migration, significantly reduced the numbers of T lymphocytes in the lung of mice. Compared with wild-type mice, Tnfaip8−/−Tipe2−/− mice also developed more severe infection with more pathogens entering blood circulation upon nasal Streptococcus pneumoniae challenge. Single-cell RNA-sequencing analysis revealed that TIPE proteins selectively affected mucosal homing of a unique subpopulation of T cells, called “T cells-2”, which expressed high levels of Ccr9, Tcf7, and Rag1/2 genes. TNFAIP8 and TIPE2 appeared to have overlapping functions since deficiency in both yielded the strongest phenotype. These data demonstrate that TIPE family of proteins are crucial for lung mucosal immunity. Strategies targeting TIPE proteins may help develop mucosal vaccines or treat inflammatory diseases of the lung.
Collapse
|
29
|
Wu J, Ning Y, Tan L, Chen Y, Huang X, Zhuo Y. Characteristics of the vaginal microbiome in women with premature ovarian insufficiency. J Ovarian Res 2021; 14:172. [PMID: 34879874 PMCID: PMC8655991 DOI: 10.1186/s13048-021-00923-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/18/2021] [Indexed: 11/30/2022] Open
Abstract
PURPOSE To investigate the relationship between vaginal microbial community structure and premature ovarian insufficiency (POI). METHODS Twenty-eight women with POI and 12 healthy women were recruited at Shenzhen Maternity and Child Healthcare Hospital between August and September 2020. Blood samples were collected for glucose tests and detection of sex hormone levels and vaginal secretions were collected for microbial group determination. Vaginal microbial community profiles were analysed by 16S rRNA gene sequencing using the Illumina MiSeq system (Illumina Inc., San Diego, CA, USA). RESULTS Compared to the controls, the serum levels of follicle-stimulating hormone, luteinizing hormone, testosterone, and the follicle-stimulating hormone/luteinizing hormone ratio, significantly increased, and oestradiol and anti-Müllerian hormone levels significantly decreased in women with POI. Higher weighted UniFrac values were observed in women with POI than in healthy women. Bacteria in the genera Lactobacillus, Brevundimonas, and Odoribacter were more abundant in the microbiomes of healthy women, while the quantity of bacteria in the genus Streptococcus was significantly increased in the microbiomes of women with POI. Moreover, these differences in microbes in women with POI were closely related to follicle-stimulating hormone, luteinizing hormone, oestradiol, and anti-Müllerian hormone levels and to the follicle-stimulating hormone/luteinizing hormone ratio. CONCLUSIONS Women with POI had altered vaginal microbial profiles compared to healthy controls. The alterations in their microbiomes were associated with serum hormone levels. These results will improve our understanding of the vaginal microbial community structure in women with POI. TRIAL REGISTRATION CHICTR, ChiCTR2000029576 . Registered 3 August 2020 - Retrospectively registered, https://www.chictr.org.cn/showproj.aspx?proj=48844 .
Collapse
Affiliation(s)
- Jiaman Wu
- Department of Chinese Medicine, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, 518028, China
| | - Yan Ning
- Department of Chinese Medicine, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, 518028, China
| | - Liya Tan
- Department of Chinese Medicine, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, 518028, China
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yan Chen
- Department of Chinese Medicine, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, 518028, China
| | - Xingxian Huang
- Department of Acupuncture and Moxibustion, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Yuanyuan Zhuo
- Department of Acupuncture and Moxibustion, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China.
| |
Collapse
|
30
|
Wang Y, Xia L, Wang G, Lu H, Wang H, Luo S, Zhang T, Gao S, Huang J, Min X. Subcutaneous immunization with the fusion protein ΔA146Ply-SP0148 confers protection against Streptococcus pneumoniae infection. Microb Pathog 2021; 162:105325. [PMID: 34848296 DOI: 10.1016/j.micpath.2021.105325] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 11/19/2021] [Accepted: 11/26/2021] [Indexed: 12/30/2022]
Abstract
Pneumococcal SP0148 and pneumolysin (Ply) derivatives are important vaccine candidates. SP0148 is a conserved lipoprotein with high immunogenicity produced by Streptococcus pneumoniae. We have previously demonstrated that SP0148 can confer protection against fatal infections caused by S. pneumoniae. ΔA146Ply is a noncytotoxic mutant of Ply that retains the TLR4 agonistic effect and has mucosal and subcutaneous adjuvant activities suggested to induce protective immunity against S. pneumoniae infection. In this study, we constructed the fusion protein ΔA146Ply-SP0148, composed of ΔA146Ply and SP0148, and evaluated the immunoprotective effect of the fusion protein. When mice were subcutaneously immunized with the fusion protein ΔA146Ply-SP0148, high levels of anti-ΔA146Ply and anti-SP0148 IgG antibodies were induced in the serum. Specific antibodies can bind to a variety of different serotypes of S. pneumoniae. Compared with mice immunized with ΔA146Ply and SP0148 alone, mice immunized subcutaneously with the fusion protein ΔA146Ply-SP0148 with Al(OH)3 had a higher survival rate when challenged by a lethal dose of S. pneumoniae, and they also had significantly lower lung bacterial loads and milder lung inflammation. In addition, mice immunized subcutaneously with the fusion protein ΔA146Ply-SP0148 stimulated strong Th1, Th2, and Th17 cell responses. In summary, these results suggest that subcutaneous immunization with the ΔA146Ply-SP0148 fusion protein can protect mice against fatal pneumococcal infection and lung infection. The fusion protein ΔA146ply-SP0148 can be a new pneumococcal vaccine target.
Collapse
Affiliation(s)
- Yao Wang
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Lingyin Xia
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Guangli Wang
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Huifang Lu
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Hui Wang
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Shilu Luo
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Tao Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Song Gao
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jian Huang
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| | - Xun Min
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
31
|
He SWJ, van de Garde MDB, Pieren DKJ, Poelen MCM, Voß F, Abdullah MR, Hammerschmidt S, van Els CACM. Diminished Pneumococcal-Specific CD4+ T-Cell Response is Associated With Increased Regulatory T Cells at Older Age. FRONTIERS IN AGING 2021; 2:746295. [PMID: 35822055 PMCID: PMC9261371 DOI: 10.3389/fragi.2021.746295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022]
Abstract
Respiratory infection caused by Streptococcus pneumoniae is a leading cause of morbidity and mortality in older adults. Acquired CD4+ T cell mechanism are essential for the protection against colonization and subsequent development of infections by S. pneumoniae. In this study, we hypothesized that age-related changes within the CD4+ T-cell population compromise CD4+ T-cell specific responses to S. pneumoniae, thereby contributing to increased susceptibility at older age. To this end, we interrogated the CD4+ T-cell response against the immunogenic pneumococcal protein AliB, part of the unique oligopeptide ABC transporter system responsible for the uptake of nutrients for the bacterium and crucial for the development of pneumococcal meningitis, in healthy young and older adults. Specifically, proliferation of CD4+ T cells as well as concomitant cytokine profiles and phenotypic markers implied in immunosenescence were studied. Older adults showed decreased AliB-induced CD4+ T-cell proliferation that is associated with an increased frequency of regulatory T cells and lower levels of active CD25+CD127+CTLA-4−TIGIT-CD4+T cells. Additionally, levels of pro-inflammatory cytokines IFNy and IL-17F were decreased at older age. Our findings indicate that key features of a pneumococcal-specific CD4+ T-cell immune response are altered at older age, which may contribute to enhanced susceptibility for pneumococcal infections.
Collapse
Affiliation(s)
- Samantha W J He
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Martijn D B van de Garde
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Daan K J Pieren
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Martien C M Poelen
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Franziska Voß
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute of Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Mohammed R Abdullah
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute of Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Interfaculty Institute of Genetics and Functional Genomics, Center for Functional Genomics of Microbes, University of Greifswald, Greifswald, Germany
| | - Cécile A C M van Els
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| |
Collapse
|
32
|
Darden DB, Dong X, Brusko MA, Kelly L, Fenner B, Rincon JC, Dirain ML, Ungaro R, Nacionales DC, Gauthier M, Kladde M, Brusko TM, Bihorac A, Moore FA, Loftus T, Bacher R, Moldawer LL, Mohr AM, Efron PA. A Novel Single Cell RNA-seq Analysis of Non-Myeloid Circulating Cells in Late Sepsis. Front Immunol 2021; 12:696536. [PMID: 34484194 PMCID: PMC8415415 DOI: 10.3389/fimmu.2021.696536] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/30/2021] [Indexed: 12/11/2022] Open
Abstract
Background With the successful implementation of the Surviving Sepsis Campaign guidelines, post-sepsis in-hospital mortality to sepsis continues to decrease. Those who acutely survive surgical sepsis will either rapidly recover or develop a chronic critical illness (CCI). CCI is associated with adverse long-term outcomes and 1-year mortality. Although the pathobiology of CCI remains undefined, emerging evidence suggests a post-sepsis state of pathologic myeloid activation, inducing suboptimal lymphopoiesis and erythropoiesis, as well as downstream leukocyte dysfunction. Our goal was to use single-cell RNA sequencing (scRNA-seq) to perform a detailed transcriptomic analysis of lymphoid-derived leukocytes to better understand the pathology of late sepsis. Methods A mixture of whole blood myeloid-enriched and Ficoll-enriched peripheral blood mononuclear cells from four late septic patients (post-sepsis day 14-21) and five healthy subjects underwent Cellular Indexing of Transcriptomes and Epitopes by Sequencing (CITE-seq). Results We identified unique transcriptomic patterns for multiple circulating immune cell subtypes, including B- and CD4+, CD8+, activated CD4+ and activated CD8+ T-lymphocytes, as well as natural killer (NK), NKT, and plasmacytoid dendritic cells in late sepsis patients. Analysis demonstrated that the circulating lymphoid cells maintained a transcriptome reflecting immunosuppression and low-grade inflammation. We also identified transcriptomic differences between patients with bacterial versus fungal sepsis, such as greater expression of cytotoxic genes among CD8+ T-lymphocytes in late bacterial sepsis. Conclusion Circulating non-myeloid cells display a unique transcriptomic pattern late after sepsis. Non-myeloid leukocytes in particular reveal a host endotype of inflammation, immunosuppression, and dysfunction, suggesting a role for precision medicine-guided immunomodulatory therapy.
Collapse
Affiliation(s)
- Dijoia B Darden
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Xiaoru Dong
- Department of Biomedical Engineering, University of Florida College of Medicine, Gainesville, FL, United States
| | - Maigan A Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Lauren Kelly
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Brittany Fenner
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Jaimar C Rincon
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Marvin L Dirain
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Ricardo Ungaro
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Dina C Nacionales
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Marie Gauthier
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL, United States
| | - Michael Kladde
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL, United States
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Azra Bihorac
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, United States
| | - Frederick A Moore
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Tyler Loftus
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Rhonda Bacher
- Department of Biostatistics, University of Florida, Gainesville, FL, United States
| | - Lyle L Moldawer
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Alicia M Mohr
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Philip A Efron
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
33
|
Application of the MISTEACHING(S) disease susceptibility framework to Actinobacillus pleuropneumoniae to identify research gaps: an exemplar of a veterinary pathogen. Anim Health Res Rev 2021; 22:120-135. [PMID: 34275511 DOI: 10.1017/s1466252321000074] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Historically, the MISTEACHING (microbiome, immunity, sex, temperature, environment, age, chance, history, inoculum, nutrition, genetics) framework to describe the outcome of host-pathogen interaction, has been applied to human pathogens. Here, we show, using Actinobacillus pleuropneumoniae as an exemplar, that the MISTEACHING framework can be applied to a strict veterinary pathogen, enabling the identification of major research gaps, the formulation of hypotheses whose study will lead to a greater understanding of pathogenic mechanisms, and/or improved prevention/therapeutic measures. We also suggest that the MISTEACHING framework should be extended with the inclusion of a 'strain' category, to become MISTEACHINGS. We conclude that the MISTEACHINGS framework can be applied to veterinary pathogens, whether they be bacteria, fungi, viruses, or parasites, and hope to stimulate others to use it to identify research gaps and to formulate hypotheses worthy of study with their own pathogens.
Collapse
|
34
|
Exploring the Ability of Meningococcal Vaccines to Elicit Mucosal Immunity: Insights from Humans and Mice. Pathogens 2021; 10:pathogens10070906. [PMID: 34358056 PMCID: PMC8308890 DOI: 10.3390/pathogens10070906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/01/2021] [Accepted: 07/09/2021] [Indexed: 12/16/2022] Open
Abstract
Neisseria meningitidis causes a devastating invasive disease but is also a normal colonizer of the human nasopharynx. Due to the rapid progression of disease, the best tool to protect individuals against meningococcal infections is immunization. Clinical experience with polysaccharide conjugate vaccines has revealed that an ideal meningococcal vaccine must prevent both invasive disease and nasal colonization, which confers herd immunity. However, not all meningococcal vaccines are equal in their ability to prevent nasal colonization, for unknown reasons. Herein, we describe recent efforts to utilize humanized mouse models to understand the impact of different meningococcal vaccines on nasal colonization. These mice are susceptible to nasal colonization, and they become immune following live nasal infection or immunization with matched capsule-conjugate or protein-based vaccines, replicating findings from human work. We bring together insights regarding meningococcal colonization and immunity from clinical work with findings using humanized mouse models, providing new perspective into the different determinants of mucosal versus systemic immunity. Then, we use this as a framework to help focus future studies toward understanding key mechanistic aspects left unresolved, including the bacterial factors required for colonization and immune evasion, determinants of nasal mucosal protection, and characteristics of an ideal meningococcal vaccine.
Collapse
|
35
|
Grant LR, Slack MPE, Yan Q, Trzciński K, Barratt J, Sobczyk E, Appleby J, Cané A, Jodar L, Isturiz RE, Gessner BD. The epidemiologic and biologic basis for classifying older age as a high-risk, immunocompromising condition for pneumococcal vaccine policy. Expert Rev Vaccines 2021; 20:691-705. [PMID: 34233558 DOI: 10.1080/14760584.2021.1921579] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Immunosenescence is a normal biologic process involving deterioration of protective immune responses. Consequently, older adults experience increased risk of infectious diseases, particularly pneumonia, and its leading bacterial cause, Streptococcus pneumoniae. Pneumococcal vaccine recommendations are often limited to adults with specific medical conditions despite similar disease risks among older adults due to immunosenescence. AREAS COVERED This article reviews epidemiologic, biologic, and clinical evidence supporting the consideration of older age due to immunosenescence as an immunocompromising condition for the purpose of pneumococcal vaccine policy and the role vaccination can play in healthy aging. EXPERT OPINION Epidemiologic and biologic evidence suggest that pneumococcal disease risk increases with age and is comparable for healthy older adults and younger adults with immunocompromising conditions. Because immunocompromising conditions are already indicated for pneumococcal conjugate vaccines (PCVs), a comprehensive public health strategy would also recognize immunosenescence. Moreover, older persons should be vaccinated before reaching the highest risk ages, consistent with the approach for other immunocompromising conditions. To facilitate PCV use among older adults, vaccine technical committees (VTCs) could classify older age as an immunocompromising condition based on the process of immunosenescence. With global aging, VTCs will need to consider immunosenescence and vaccine use during healthy aging.
Collapse
Affiliation(s)
- Lindsay R Grant
- Vaccines Medical Development & Scientific/Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| | - Mary P E Slack
- School of Medicine, Griffith University Gold Coast Campus, Australia
| | - Qi Yan
- Vaccines Medical Development & Scientific/Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| | - Krzysztof Trzciński
- Department of Pediatric Immunology and Infectious Diseases, Wilhelmina's Children Hospital, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Jane Barratt
- International Federation on Ageing, Toronto, Ontario, Canada
| | | | - James Appleby
- The Gerontological Society of America, Washington, D.C., USA
| | - Alejandro Cané
- Vaccines Medical Development & Scientific/Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| | - Luis Jodar
- Vaccines Medical Development & Scientific/Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| | - Raul E Isturiz
- Vaccines Medical Development & Scientific/Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| | - Bradford D Gessner
- Vaccines Medical Development & Scientific/Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| |
Collapse
|
36
|
Huang ST, Yu TM, Chuang YW, Chen CH, Wu MJ, Wang IK, Li CY, Lin CL, Kao CH. Pneumococcal pneumonia in adult hospitalised solid organ transplant recipients: Nationwide, population-based surveillance. Int J Clin Pract 2021; 75:e14126. [PMID: 33638887 DOI: 10.1111/ijcp.14126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 02/26/2021] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Pneumococcal disease poses a burden to the community in high risk population. Most early studies focused on invasive pneumococcal disease. However, the epidemiology of pneumococcal pneumonia (PP) requiring hospitalisation in solid organ transplant recipients (SOTRs) is poorly defined. METHODS We conducted a retrospective cohort study (January 1, 2000 and December 31, 2012) to evaluate the risk of PP requiring hospitalisation in SOTRs. SOTRs and non-SOT cohorts, propensity score-matched at a 1:1 ratio for age, sex, index date and underlying comorbidities, were identified from the National Health Insurance Research Database. RESULTS Each cohort consisted of 7507 patients. In the SOT cohort, 26 episodes of PP requiring hospitalisation were identified (incidence rate of 52.4 per 100,000 person-years). The risk of PP requiring hospitalisation in the SOT cohort was 1.50 times greater than in the non-SOT cohort [adjusted hazard ratio: 1.50, 95% confidence interval = 1.31-1.71, P < .001]. The nested case control study identified older age, kidney transplant, and concomitant chronic obstructive pulmonary disease, chronic kidney disease and heart failure as predictors of PP requiring hospitalisation in the SOT cohort. The highest risk period for PP requiring hospitalisation occurred within the first year of transplantation (36.47 per 1000 patients). Amongst kidney transplant recipients, patients with PP requiring hospitalisation exhibited higher cumulative incidences of graft failure than those without PP (log-rank test: P value = .004). CONCLUSIONS SOTRs are at risk of PP requiring hospitalisation with its attendant morbidity. Strategies to reduce risk of PP requiring hospitalisation using preventive vaccinations warrant further study.
Collapse
Affiliation(s)
- Shih-Ting Huang
- Division of Nephrology, Taichung Veterans General Hospital, Taichung, Taiwan
- Graduate Institute of Public Health, China Medical University, Taichung, Taiwan
| | - Tung-Min Yu
- Division of Nephrology, Taichung Veterans General Hospital, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan
| | - Ya-Wen Chuang
- Division of Nephrology, Taichung Veterans General Hospital, Taichung, Taiwan
- Graduate Institute of Public Health, China Medical University, Taichung, Taiwan
| | - Cheng-Hsu Chen
- Division of Nephrology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ming-Ju Wu
- Division of Nephrology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - I-Kuan Wang
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan
- Division of Nephrology, China Medical University Hospital, Taichung, Taiwan
| | - Chi-Yuan Li
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan
- Division of Anesthesiology, China Medical University Hospital, Taichung, Taiwan
| | - Cheng-Li Lin
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
| | - Chia-Hung Kao
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung, Taiwan
- Department of Nuclear Medicine and PET Center, China Medical University, Taichung, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
- Center of Augmented Intelligence in Healthcare, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
37
|
Barker KA, Etesami NS, Shenoy AT, Arafa EI, Lyon de Ana C, Smith NM, Martin IM, Goltry WN, Barron AM, Browning JL, Kathuria H, Belkina AC, Guillon A, Zhong X, Crossland NA, Jones MR, Quinton LJ, Mizgerd JP. Lung-resident memory B cells protect against bacterial pneumonia. J Clin Invest 2021; 131:e141810. [PMID: 34060477 DOI: 10.1172/jci141810] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 04/14/2021] [Indexed: 12/22/2022] Open
Abstract
Lung-resident memory B cells (BRM cells) are elicited after influenza infections of mice, but connections to other pathogens and hosts - as well as their functional significance - have yet to be determined. We postulate that BRM cells are core components of lung immunity. To test this, we examined whether lung BRM cells are elicited by the respiratory pathogen pneumococcus, are present in humans, and are important in pneumonia defense. Lungs of mice that had recovered from pneumococcal infections did not contain organized tertiary lymphoid organs, but did have plasma cells and noncirculating memory B cells. The latter expressed distinctive surface markers (including CD69, PD-L2, CD80, and CD73) and were poised to secrete antibodies upon stimulation. Human lungs also contained B cells with a resident memory phenotype. In mice recovered from pneumococcal pneumonia, depletion of PD-L2+ B cells, including lung BRM cells, diminished bacterial clearance and the level of pneumococcus-reactive antibodies in the lung. These data define lung BRM cells as a common feature of pathogen-experienced lungs and provide direct evidence of a role for these cells in pulmonary antibacterial immunity.
Collapse
Affiliation(s)
| | | | | | | | | | - Nicole Ms Smith
- Pulmonary Center.,Department of Pathology and Laboratory Medicine, and
| | | | | | | | | | | | - Anna C Belkina
- Pulmonary Center.,Department of Pathology and Laboratory Medicine, and.,Flow Cytometry Core Facility, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Antoine Guillon
- Pulmonary Center.,Centre Hospitalier Régional Universitaire de (CHRU) de Tours, Service de Médecine Intensive Réanimation, INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, University of Tours, Tours, France
| | | | | | | | - Lee J Quinton
- Pulmonary Center.,Department of Microbiology.,Department of Medicine.,Department of Pathology and Laboratory Medicine, and
| | - Joseph P Mizgerd
- Pulmonary Center.,Department of Microbiology.,Department of Medicine.,Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
38
|
Ramos-Sevillano E, Ercoli G, Felgner P, Ramiro de Assis R, Nakajima R, Goldblatt D, Heyderman RS, Gordon SB, Ferreira DM, Brown JS. Preclinical Development of Virulence-attenuated Streptococcus pneumoniae Strains Able to Enhance Protective Immunity against Pneumococcal Infection. Am J Respir Crit Care Med 2021; 203:1037-1041. [PMID: 33332997 DOI: 10.1164/rccm.202011-4161le] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
| | | | | | | | - Rie Nakajima
- University of California Irvine Irvine, California
| | | | | | - Stephen B Gordon
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme Blantyre, Malawi and
| | | | | |
Collapse
|
39
|
Camelo IY, Mwananyanda LM, Thea DM, Seidenberg P, Gill CJ, Weinstein JR. A Tale of 2 Pneumos: The Impact of Human Immunodeficiency Virus Exposure or Infection Status on Pediatric Nasopharyngeal Carriage of Streptococcus pneumoniae and Pneumocystis jiroveci: A Nested Case Control Analysis From the Pneumonia Etiology Research In Child Health Study. Clin Infect Dis 2021; 72:1033-1041. [PMID: 32342105 DOI: 10.1093/cid/ciaa164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/10/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The majority of pediatric human immunodeficiency virus (HIV) cases in Africa reflect maternal-to-child transmission. HIV exposed but uninfected (HEU) children have increased rates of morbidity and mortality when compared to HIV unexposed and uninfected (HUU) children. The mechanisms behind these unexpected trends are only partially understood but could be explained by the differences in the immune response to infections triggered by an altered immune system state. METHODS Using quantitative reverse transcription polymerase chain reaction, we compared the nasopharyngeal carriage prevalence and density of Streptococcus pneumoniae (SP) and Pneumocystis jirovecii (PJ) between children living with HIV and HEU or HUU cases (pneumonia) and controls (without pneumonia). RESULTS The cohort included 1154 children (555 cases and 599 matched controls). The SP carriage prevalence rates were similar between cases and controls. Among SP carriers with pneumonia, carriage density was increased among children living with HIV, versus HEU or HUU children (15.8, 4.7, and 3.6 × 105 copies/mL, respectively). The rate of PJ carriage was significantly higher among children living with HIV than among HEU and HUU children (31%, 15%, and 10%, respectively; P < .05), as was carriage density (63.9, 20.9, and 4.8 × 103 copies/mL, respectively; P < .05). CONCLUSIONS Carriage prevalences and densities for SP and PJ show different kinetics in terms of their relationship with HIV exposure and clinical status, particularly for Pneumocystis jirovecii. This supports the theory that the increased morbidity and mortality observed among HEU children may reflect deficits not just in humoral immunity but in cell-mediated immunity as well.
Collapse
Affiliation(s)
- Ingrid Y Camelo
- Pediatric Infectious Diseases Department, Boston University Medical Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Lawrence M Mwananyanda
- Department of Global Health, Boston University School of Public Health, Boston, Massachusetts, USA.,Right to Care Zambia, Lusaka, Zambia
| | - Donald M Thea
- Department of Global Health, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Philip Seidenberg
- Department of Emergency Medicine, University of New Mexico, Albuquerque, New Mexico, USA
| | - Christopher J Gill
- Department of Global Health, Boston University School of Public Health, Boston, Massachusetts, USA
| | - John R Weinstein
- Department of Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
40
|
Ferrara F, Rial A, Suárez N, Chabalgoity JA. Polyvalent Bacterial Lysate Protects Against Pneumonia Independently of Neutrophils, IL-17A or Caspase-1 Activation. Front Immunol 2021; 12:562244. [PMID: 33981296 PMCID: PMC8108696 DOI: 10.3389/fimmu.2021.562244] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 03/18/2021] [Indexed: 11/28/2022] Open
Abstract
Polyvalent bacterial lysates have been in use for decades for prevention and treatment of respiratory infections with reported clinical benefits. However, besides claims of broad immune activation, the mode of action is still a matter of debate. The lysates, formulated with the main bacterial species involved in respiratory infections, are commonly prepared by chemical or mechanical disruption of bacterial cells, what is believed influences the biological activity of the product. Here, we prepared two polyvalent lysates with the same composition but different method of bacterial cell disruption and evaluated their biological activity in a comparative fashion. We found that both bacterial lysates induce NF-kB activation in a MyD88 dependent manner, suggesting they work as TLR agonists. Further, we found that a single intranasal dose of any of the two lysates, is sufficient to protect against pneumococcal pneumonia, suggesting that they exert similar biological activity. We have previously shown that protection against pneumococcal pneumonia can also be induced by prior S. pneumoniae sub lethal infection or therapeutic treatment with a TLR5 agonist. Protection in those cases depends on neutrophil recruitment to the lungs, and can be associated with increased local expression of IL-17A. Here, we show that bacterial lysates exert protection against pneumococcal pneumonia independently of neutrophils, IL-17A or Caspase-1/11 activation, suggesting the existence of redundant mechanisms of protection. Trypsin-treated lysates afford protection to the same extent, suggesting that just small peptides suffice to exert the protective effect or that the molecules responsible for the protective effect are not proteins. Understanding the mechanism of action of bacterial lysates and deciphering the active components shall allow redesigning them with more precisely defined formulations and expanding their range of action.
Collapse
Affiliation(s)
- Florencia Ferrara
- Laboratory for Vaccine Research, Departamento de Desarrollo Biotecnológico, Facultad de Medicina, Instituto de Higiene, Montevideo, Uruguay
| | - Analía Rial
- Laboratory for Vaccine Research, Departamento de Desarrollo Biotecnológico, Facultad de Medicina, Instituto de Higiene, Montevideo, Uruguay
| | - Norma Suárez
- Laboratory for Vaccine Research, Departamento de Desarrollo Biotecnológico, Facultad de Medicina, Instituto de Higiene, Montevideo, Uruguay
| | - José Alejandro Chabalgoity
- Laboratory for Vaccine Research, Departamento de Desarrollo Biotecnológico, Facultad de Medicina, Instituto de Higiene, Montevideo, Uruguay
| |
Collapse
|
41
|
Cripps AW, Folaranmi T, Johnson KD, Musey L, Niederman MS, Buchwald UK. Immunogenicity following revaccination or sequential vaccination with 23-valent pneumococcal polysaccharide vaccine (PPSV23) in older adults and those at increased risk of pneumococcal disease: a review of the literature. Expert Rev Vaccines 2021; 20:257-267. [PMID: 33567914 DOI: 10.1080/14760584.2021.1889374] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction: Immunogenicity studies evaluating sequential administration of pneumococcal conjugate vaccine (PCV) followed by 23-valent pneumococcal polysaccharide vaccine (PPSV23) or revaccination with PPSV23 have raised concerns that PPSV23 may not elicit higher antibody levels than those measured following PCV or first PPSV23 dose.Areas covered: Recent literature was evaluated for evidence of blunted immune response (hyporesponsiveness), focusing on studies using adequate intervals between doses in accordance with vaccination recommendations. In eight of nine studies that evaluated revaccination with PPSV23 at an interval of ≥5 years after the previous dose, immunoglobulin G geometric mean concentrations and/or opsonophagocytic assay geometric mean titers for most serotypes increased from pre- to post-repeat vaccination and were comparable between repeat and primary vaccination groups post-vaccination. In seven studies in which PPSV23 was administered after PCVs (8 weeks to 1 year apart), responses to PPSV23 were comparable to those seen after initial PCV dose for shared vaccine serotypes. Studies in which PCVs were administered after PPSV23 were not evaluated.Expert opinion: Published data suggest immune responses following repeat vaccination with PPSV23, or sequential PCV/PPSV23 vaccination, are robust, without evidence of hyporesponsiveness. PPSV23 vaccination of at-risk adults is essential to ensure broad protection against all 23 vaccine serotypes.
Collapse
Affiliation(s)
- Allan W Cripps
- Mucosal Immunology Research Group, Menzies Health Institute and School of Medicine, Griffith University, Gold Coast Campus, Southport QLD, Australia
| | | | | | | | | | | |
Collapse
|
42
|
Sputum Proteome Signatures of Mechanically Ventilated Intensive Care Unit Patients Distinguish Samples with or without Anti-pneumococcal Activity. mSystems 2021; 6:6/2/e00702-20. [PMID: 33653939 PMCID: PMC8546979 DOI: 10.1128/msystems.00702-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Mechanically ventilated patients are at risk of contracting pneumonia. Therefore, these patients often receive prophylactic systemic antimicrobial therapy. Intriguingly however, a previous study showed that antimicrobial activity in bronchoalveolar aspirates (here referred to as “sputa”) from ventilated patients was only partially explained by antibiotic therapy. Here we report that sputa from these patients presented distinct proteome signatures depending on the presence or absence of antimicrobial activity. Moreover, we show that the same distinction applied to antibodies against Streptococcus pneumoniae, which is a major causative agent of pneumonia. Specifically, the investigated sputa that inhibited growth of S. pneumoniae, while containing subinhibitory levels of the antibiotic cefotaxime, presented elevated levels of proteins implicated in innate immune defenses, including complement and apolipoprotein-associated proteins. In contrast, S. pneumoniae-inhibiting sputa with relatively high cefotaxime concentrations or noninhibiting sputa contained higher levels of proteins involved in inflammatory responses, such as neutrophil elastase-associated proteins. In an immunoproteomics analysis, 18 out of 55 S. pneumoniae antigens tested showed significantly increased levels of IgGs in inhibiting sputa. Hence, proteomics and immunoproteomics revealed elevated levels of antimicrobial host proteins or S. pneumoniae antigen-specific IgGs in pneumococcal growth-inhibiting sputa, thus explaining their anti-pneumococcal activity. IMPORTANCE Respiratory pathogens like Streptococcus pneumoniae can cause severe pneumonia. Nonetheless, mechanically ventilated intensive care patients, who have a high risk of contracting pneumonia, rarely develop pneumococcal pneumonia. This suggests the presence of potentially protective antimicrobial agents in their lung environment. Our present study shows for the first time that bronchoalveolar aspirates, “sputa,” of ventilated patients in a Dutch intensive care unit were characterized by three distinct groups of proteome abundance signatures that can explain their anti-pneumococcal activity. Importantly, this anti-pneumococcal sputum activity was related either to elevated levels of antimicrobial host proteins or to antibiotics and S. pneumoniae-specific antibodies. Further, the sputum composition of some patients changed over time. Therefore, we conclude that our study may provide a novel tool to measure changes that are indicative of infection-related conditions in the lungs of mechanically ventilated patients.
Collapse
|
43
|
Scott NR, Mann B, Tuomanen EI, Orihuela CJ. Multi-Valent Protein Hybrid Pneumococcal Vaccines: A Strategy for the Next Generation of Vaccines. Vaccines (Basel) 2021; 9:209. [PMID: 33801372 PMCID: PMC8002124 DOI: 10.3390/vaccines9030209] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/15/2022] Open
Abstract
Streptococcus pneumoniae (Spn) is a bacterial pathogen known to colonize the upper respiratory tract and cause serious opportunistic diseases such as pneumonia, bacteremia, sepsis and meningitis. As a consequence, millions of attributable deaths occur annually, especially among infants, the elderly and immunocompromised individuals. Although current vaccines, composed of purified pneumococcal polysaccharide in free form or conjugated to a protein carrier, are widely used and have been demonstrated to be effective in target groups, Spn has continued to colonize and cause life-threatening disease in susceptible populations. This lack of broad protection highlights the necessity of improving upon the current "gold standard" pneumococcal vaccines to increase protection both by decreasing colonization and reducing the incidence of sterile-site infections. Over the past century, most of the pneumococcal proteins that play an essential role in colonization and pathogenesis have been identified and characterized. Some of these proteins have the potential to serve as antigens in a multi-valent protein vaccine that confers capsule independent protection. This review seeks to summarize the benefits and limitations of the currently employed vaccine strategies, describes how leading candidate proteins contribute to pneumococcal disease development, and discusses the potential of these proteins as protective antigens-including as a hybrid construct.
Collapse
Affiliation(s)
- Ninecia R. Scott
- Department of Microbiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Beth Mann
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (B.M.); (E.I.T.)
| | - Elaine I. Tuomanen
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (B.M.); (E.I.T.)
| | - Carlos J. Orihuela
- Department of Microbiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| |
Collapse
|
44
|
Ercoli G, Ramos-Sevillano E, Nakajima R, de Assis RR, Jasinskas A, Goldblatt D, Felgner P, Weckbecker G, Brown J. The Influence of B Cell Depletion Therapy on Naturally Acquired Immunity to Streptococcus pneumoniae. Front Immunol 2021; 11:611661. [PMID: 33584691 PMCID: PMC7876223 DOI: 10.3389/fimmu.2020.611661] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/14/2020] [Indexed: 02/03/2023] Open
Abstract
The anti-CD20 antibody Rituximab to deplete CD20+ B cells is an effective treatment for rheumatoid arthritis and B cell malignancies, but is associated with an increased incidence of respiratory infections. Using mouse models we have investigated the consequences of B cell depletion on natural and acquired humoral immunity to Streptococcus pneumoniae. B cell depletion of naïve C57Bl/6 mice reduced natural IgM recognition of S. pneumoniae, but did not increase susceptibility to S. pneumoniae pneumonia. ELISA and flow cytometry assays demonstrated significantly reduced IgG and IgM recognition of S. pneumoniae in sera from mice treated with B cell depletion prior to S. pneumoniae nasopharyngeal colonization compared to untreated mice. Colonization induced antibody responses to protein rather than capsular antigen, and when measured using a protein array B cell depletion prior to colonization reduced serum levels of IgG to several protein antigens. However, B cell depleted S. pneumoniae colonized mice were still partially protected against both lung infection and septicemia when challenged with S. pneumoniae after reconstitution of their B cells. These data indicate that although B cell depletion markedly impairs antibody recognition of S. pneumoniae in colonized mice, some protective immunity is maintained, perhaps mediated by cellular immunity.
Collapse
MESH Headings
- Animals
- Antibodies, Bacterial/blood
- B-Lymphocytes/drug effects
- B-Lymphocytes/immunology
- Disease Models, Animal
- Female
- Host-Pathogen Interactions
- Immunity, Cellular
- Immunity, Humoral
- Immunity, Innate
- Immunoglobulin G/blood
- Immunoglobulin M/blood
- Immunologic Factors/pharmacology
- Lymphocyte Depletion
- Mice, Inbred C57BL
- Pneumonia, Pneumococcal/blood
- Pneumonia, Pneumococcal/immunology
- Pneumonia, Pneumococcal/microbiology
- Pneumonia, Pneumococcal/prevention & control
- Rituximab/pharmacology
- Streptococcus pneumoniae/immunology
- Streptococcus pneumoniae/pathogenicity
- Mice
Collapse
Affiliation(s)
- Giuseppe Ercoli
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Division of Medicine, University College Medical School, Rayne Institute, London, United Kingdom
| | - Elisa Ramos-Sevillano
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Division of Medicine, University College Medical School, Rayne Institute, London, United Kingdom
| | - Rie Nakajima
- Vaccine Research and Development Center, Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, United States
| | - Rafael Ramiro de Assis
- Vaccine Research and Development Center, Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, United States
| | - Algis Jasinskas
- Vaccine Research and Development Center, Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, United States
| | - David Goldblatt
- Department of Immunobiology, UCL Great Ormond Street Institute of Child Health, NIHR Biomedical Research Centre, London, United Kingdom
| | - Philip Felgner
- Vaccine Research and Development Center, Department of Physiology and Biophysics, University of California Irvine, Irvine, CA, United States
| | - Gisbert Weckbecker
- Novartis Institute for BioMedical Research, Novartis, Basel, Switzerland
| | - Jeremy Brown
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Division of Medicine, University College Medical School, Rayne Institute, London, United Kingdom
| |
Collapse
|
45
|
Weng CC, Chao CY, Wu ST, Tsou PH, Chen WT, Li BR, Li YK. Integration of Ni/NiO nanoparticles and a microfluidic ELISA chip to generate a sensing platform for Streptococcus pneumoniae detection. RSC Adv 2021; 11:28551-28556. [PMID: 35478579 PMCID: PMC9038132 DOI: 10.1039/d1ra04631d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/16/2021] [Indexed: 11/21/2022] Open
Abstract
Enzyme-linked immunosorbent assays (ELISAs) are tests that uses antibody recognition and enzyme catalytic activity to identify a substance, and they have been widely used as a diagnostic tool in the clinic.
Collapse
Affiliation(s)
- Chang-Ching Weng
- Department of Applied Chemistry, College of Science, Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Chien-Yu Chao
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - She-Ting Wu
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Ping-Hsien Tsou
- Department of Internal Medicine, National Taiwan University Hospital, Hsin-Chu Branch, Hsinchu, Taiwan
| | - Wei-Tin Chen
- Center for Condensed Matter Sciences, Center of Atomic Initiative for New Materials, National Taiwan University, Taipei, Taiwan
| | - Bor-Ran Li
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, Yang Ming Chiao Tung University, Hsinchu, Taiwan
- Department of Eectrical and Computer Engineering, College of Electrical and Computer Engineering, Yang Ming Chiao Tung University, Hsinchu, Taiwan
- Center for Emergent Functional Matter Science, Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Yaw-Kuen Li
- Department of Applied Chemistry, College of Science, Yang Ming Chiao Tung University, Hsinchu, Taiwan
- Center for Emergent Functional Matter Science, Yang Ming Chiao Tung University, Hsinchu, Taiwan
| |
Collapse
|
46
|
Mechanism and inhibition of Streptococcus pneumoniae IgA1 protease. Nat Commun 2020; 11:6063. [PMID: 33247098 PMCID: PMC7695701 DOI: 10.1038/s41467-020-19887-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/30/2020] [Indexed: 01/27/2023] Open
Abstract
Opportunistic pathogens such as Streptococcus pneumoniae secrete a giant metalloprotease virulence factor responsible for cleaving host IgA1, yet the molecular mechanism has remained unknown since their discovery nearly 30 years ago despite the potential for developing vaccines that target these enzymes to block infection. Here we show through a series of cryo-electron microscopy single particle reconstructions how the Streptococcus pneumoniae IgA1 protease facilitates IgA1 substrate recognition and how this can be inhibited. Specifically, the Streptococcus pneumoniae IgA1 protease subscribes to an active-site-gated mechanism where a domain undergoes a 10.0 Å movement to facilitate cleavage. Monoclonal antibody binding inhibits this conformational change, providing a direct means to block infection at the host interface. These structural studies explain decades of biological and biochemical studies and provides a general strategy to block Streptococcus pneumoniae IgA1 protease activity to potentially prevent infection. Pathogenic IgA1 metalloproteases block the initial host immune response by cleaving host IgA1. Using cryoEM, the authors here provide structural insights into the substrate recognition mechanism of Streptococcus pneumoniae IgA1 protease, and develop a protease-inhibiting antibody.
Collapse
|
47
|
Li N, Fan X, Xu M, Zhou Y, Wang B. Flu Virus Attenuates Memory Clearance of Pneumococcus via IFN-γ-Dependent Th17 and Independent Antibody Mechanisms. iScience 2020; 23:101767. [PMID: 33251497 PMCID: PMC7683269 DOI: 10.1016/j.isci.2020.101767] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 06/16/2020] [Accepted: 10/30/2020] [Indexed: 12/25/2022] Open
Abstract
Bacterial coinfection is a major cause of influenza-associated mortality. Most people have experienced infections with bacterial pathogens commonly associated with influenza A virus (IAV) coinfection before IAV exposure; however, bacterial clearance through the immunological memory response (IMR) in coinfected patients is inefficient, suggesting that the IMR to bacteria is impaired during IAV infection. Adoptive transfer of CD4+ T cells from mice that had experienced bacterial infection into IAV-infected mice revealed that memory protection against bacteria was weakened in the latter. Additionally, memory Th17 cell responses were impaired due to an IFN-γ-dependent reduction in Th17 cell proliferation and delayed migration of CD4+ T cells into the lungs. A bacterium-specific antibody-mediated memory response was also substantially reduced in coinfected mice, independently of IFN-γ. These findings provide additional perspectives on the pathogenesis of coinfection and suggest additional strategies for the treatment of defective antibacterial immunity and the design of bacterial vaccines against coinfection. Memory protection against bacteria was impaired in coinfection Memory Th17 response to bacteria was reduced by IAV-induced IFN-γ The Th17 reduction was caused by impeded Th17 proliferation and migration Bacteria-specific antibody was reduced in coinfection independent of IFN-γ
Collapse
Affiliation(s)
- Ning Li
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xin Fan
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Meiyi Xu
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Ya Zhou
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Beinan Wang
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
48
|
Orami T, Ford R, Kirkham LA, Thornton R, Corscadden K, Richmond PC, Pomat WS, van den Biggelaar AHJ, Lehmann D. Pneumococcal conjugate vaccine primes mucosal immune responses to pneumococcal polysaccharide vaccine booster in Papua New Guinean children. Vaccine 2020; 38:7977-7988. [PMID: 33121845 PMCID: PMC7684155 DOI: 10.1016/j.vaccine.2020.10.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/30/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023]
Abstract
Introduction Invasive pneumococcal disease remains a major cause of hospitalization and death in Papua New Guinean (PNG) children. We assessed mucosal IgA and IgG responses in PNG infants vaccinated with pneumococcal conjugate vaccine (PCV) followed by a pneumococcal polysaccharide vaccine (PPV) booster. Methods Infants received 7-valent PCV (7vPCV) in a 0–1–2 (neonatal) or 1–2-3-month (infant) schedule, or no 7vPCV (control). At age 9 months all children received 23-valent PPV (23vPPV). IgA and IgG to 7vPCV and non-7vPCV (1, 5, 7F, 19A) serotypes were measured in saliva collected at ages 1, 2, 3, 4, 9, 10 and 18 months (131 children, 917 samples). Correlations were studied between salivary and serum IgG at 4, 10 and 18 months. Results Salivary IgA and IgG responses overall declined in the first 9 months. Compared to non-7vPCV recipients, salivary IgA remained higher in 7vPCV recipients for serotypes 4 at 3 months, 6B at 3 months (neonatal), and 14 at 3 (neonatal), 4 and 9 months (infant); and for salivary IgG for serotypes 4 at 3, 4 and 9 months, 6B at 9 months, 14 at 4 (neonatal) and 9 months, 18C at 3, 4, and 9 (infant) months, and 23F at 4 months. Following 23vPPV, salivary 7vPCV-specific IgA and IgG increased in 7vPCV-vaccinated children but not in controls; and salivary IgA against non-PCV serotypes 5 and 7F increased in 7vPCV recipients and non-recipients. Salivary and serum IgG against 7vPCV-serotypes correlated in 7vPCV-vaccinated children at 4 and 10 months of age. Conclusions PCV may protect high-risk children against pneumococcal colonization and mucosal disease by inducing mucosal antibody responses and priming for mucosal immune memory that results in mucosal immune responses after booster PPV. Saliva can be a convenient alternative sample to serum to study PCV-induced systemic IgG responses.
Collapse
Affiliation(s)
- Tilda Orami
- Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands Province, Papua New Guinea
| | - Rebecca Ford
- Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands Province, Papua New Guinea
| | - Lea-Ann Kirkham
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia; Centre for Child Health Research, The University of Western Australia, Perth, Western Australia, Australia
| | - Ruth Thornton
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia; School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Karli Corscadden
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia
| | - Peter C Richmond
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia; Centre for Child Health Research, The University of Western Australia, Perth, Western Australia, Australia; Division of Pediatrics, School of Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - William S Pomat
- Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands Province, Papua New Guinea; Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia
| | - Anita H J van den Biggelaar
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia.
| | - Deborah Lehmann
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia; Centre for Child Health Research, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
49
|
Tuomanen EI. Perspective of a Pediatrician: Shared Pathogenesis of the Three Most Successful Pathogens of Children. Front Cell Infect Microbiol 2020; 10:585791. [PMID: 33178633 PMCID: PMC7593378 DOI: 10.3389/fcimb.2020.585791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/30/2020] [Indexed: 01/16/2023] Open
Abstract
Highly successful invasive pathogens exploit host vulnerabilities by adapting tools to co-opt highly conserved host features. This is especially true when pathogens develop ligands to hijack trafficking routes or signaling patterns of host receptors. In this context, highly successful pathogens can be grouped together by the patterns of organs infected and diseases they cause. In the case of this perspective, the focus is on the historically most successful invasive bacterial pathogens of children that cause pneumonia, sepsis and meningitis: Streptococcus pneumoniae, Haemophilus influenzae, and Neisseria meningitidis. This triad shares a ligand to bind to PAF receptor to enter host cells despite early defenses by innate immunity. All three also target laminin receptor to cross endothelial barriers using a common set of molecular tools that may prove to be a design for a cross-protective vaccine.
Collapse
Affiliation(s)
- Elaine I Tuomanen
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, United States
| |
Collapse
|
50
|
van Meel ER, Jaddoe VWV, Looman KIM, de Jongste JC, Moll HA, Duijts L. Airway bacterial carriage and childhood respiratory health: A population-based prospective cohort study. Pediatr Allergy Immunol 2020; 31:774-782. [PMID: 32524657 PMCID: PMC7587008 DOI: 10.1111/pai.13310] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/29/2020] [Accepted: 06/02/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Airway bacterial carriage might play a role in respiratory disease. We hypothesize that nasal carriage with Staphylococcus aureus or nasopharyngeal carriage with Haemophilus influenzae, Moraxella catarrhalis, and Streptococcus pneumoniae predisposes individuals to adverse respiratory health. OBJECTIVE To examine the association of early-life airway bacterial carriage with respiratory tract infections and vice versa, and of early-life airway bacterial carriage with wheezing, lung function, and asthma in later childhood. METHODS We collected upper airway swabs for bacterial culturing for S aureus, H influenzae, M catarrhalis, and H influenzae at six timepoints between the ages of 6 weeks and 6 years among 945 children participating in a population-based prospective cohort study. Information on respiratory tract infections and wheezing until age 6 years, and asthma at age 10 years was obtained by questionnaires. Lung function at age 10 years was measured by spirometry. We tested possible bidirectional associations between airway bacterial carriage and respiratory tract infections by cross-lagged models, and associations of repeatedly measured airway bacterial carriage with wheezing, lung function, and asthma by generalized estimating equations models and regression models. RESULTS Cross-lagged modeling showed that early-life airway bacterial carriage was not consistently associated with upper and lower respiratory tract infections or vice versa. Nasopharyngeal carriage with any bacteria in infancy was associated with an increased risk of wheezing (OR [95% CI]: 1.66 [1.31, 2.10]). Airway bacterial carriage was not consistently associated with school-age lung function or asthma. CONCLUSION Nasopharyngeal carriage with any bacteria is associated with wheezing, but not respiratory tract infections, asthma, or lung function.
Collapse
Affiliation(s)
- Evelien R van Meel
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Division of Respiratory Medicine and Allergology, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Vincent W V Jaddoe
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Kirsten I M Looman
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Johan C de Jongste
- Division of Respiratory Medicine and Allergology, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Henriëtte A Moll
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Liesbeth Duijts
- Division of Respiratory Medicine and Allergology, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Division of Neonatology, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|