1
|
Souod N, Bahrami F, Riazi-Rad F, Pakzad SR, Nikbin VS, Ajdary S. Robust immune responses to intranasal vaccine targeting Bordetella pertussis antigens to claudin-4 on mucosal microfold cells. Microb Pathog 2025; 204:107566. [PMID: 40220802 DOI: 10.1016/j.micpath.2025.107566] [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: 01/30/2025] [Revised: 03/23/2025] [Accepted: 04/08/2025] [Indexed: 04/14/2025]
Abstract
Despite high global vaccination rates, pertussis remains uncontrolled and is increasing in many regions. The present study aimed to develop an intranasal (I.N.) vaccine (F1S1-C-CPE) against Bordetella (B.) pertussis. This vaccine comprises the C-terminal fragment of Clostridium perfringens enterotoxin (C-CPE) fused with F1S1 antigen, which consists of the S1 subunit of pertussis toxin and the immunodominant domain of filamentous hemagglutinin (F1). C-CPE specifically targets Claudin-4 on mucosal microfold cells, which enhances immune response at the site of infection. We evaluated the immunogenicity and protective efficacy of this construct following I.N. immunization in BALB/c mice. Significant differences in serum IgG and IgA, as well as lung IgA levels were observed among mice immunized with F1S1, CPE + F1S1, F1S1-C-CPE, and PBS (p < 0.0001). Notably, the F1S1-C-CPE group exhibited significantly higher levels of IgA in both serum and lung extracts compared to the F1S1 or CPE + F1S1 groups (p < 0.05). Moreover, analysis of cell-mediated responses indicated that IFN-γ and IL-17 levels were highest in group F1S1-C-CPE, significantly exceeding those in the F1S1, F1S1+C-CPE groups (p < 0.001). Cytokine profile analysis in lung tissue post-challenge indicated the greatest fold increases in mRNA levels of IFN-γ, IL-6, IL-17, and IL-10 in group F1S1-C-CPE (p < 0.0001). The challenge results indicated that I.N. immunization with F1S1-C-CPE protein effectively controls B. pertussis infection (p < 0.0001). The results indicate the development of mucosal and systemic humoral and Th1/Th17 responses, which are critical in controlling infection. Overall, our findings highlight the potential of the F1S1-C-CPE I.N. vaccine as a powerful tool in combating pertussis.
Collapse
Affiliation(s)
- Negar Souod
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran; Pasteur Institute of Iran, Tehran, Iran.
| | - Fariborz Bahrami
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran.
| | - Farhad Riazi-Rad
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran.
| | - Saeed Reza Pakzad
- Vaccine Potency and Standardization Section, Food and Drug Control Laboratory (FDCL), Ministry of Health and Medical Education, Tehran, Iran.
| | | | - Soheila Ajdary
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
2
|
Kumar A, Johnson D, Bukowski A, Noto MJ, Carbonetti NH. Interferon lambda signaling in neutrophils enhances the pathogenesis of Bordetella pertussis infection. J Leukoc Biol 2025; 117:qiae202. [PMID: 39302155 PMCID: PMC11879762 DOI: 10.1093/jleuko/qiae202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/31/2024] [Accepted: 09/18/2024] [Indexed: 09/22/2024] Open
Abstract
Interferon lambda plays diverse roles in bacterial infections. Previously, we showed that interferon lambda is induced in the lungs of Bordetella pertussis-infected adult mice and exacerbates inflammation. Here, we report that mice lacking the interferon lambda receptor 1 specifically on neutrophils (MRP8creIFNLR1fl/fl mice) exhibit reduced lung bacterial load and inflammation compared to wild-type mice during B. pertussis infection. In B. pertussis-infected wild-type mice, lung type I and III IFN responses were higher than in MRP8creIFNLR1fl/fl mice, correlating with increased lung inflammatory pathology. There was an increased proportion of interferon gamma-producing neutrophils in the lungs of MRP8creIFNLR1fl/fl mice compared to wild-type mice. IFNLR1-/- neutrophils incubated with B. pertussis exhibited higher killing compared to wild-type neutrophils. Treatment of wild-type neutrophils with interferon lambda further decreased their bacterial killing capacity and treatment of wild-type mice with interferon lambda increased lung bacterial loads. Contributing to the differential killing, we found that IFNLR1-/- neutrophils exhibit higher levels of reactive oxygen species, myeloperoxidase, matrix metalloproteinase-9 activity, neutrophil extracellular traps, and interferon gamma secretion than wild-type neutrophils, and inhibiting NADPH oxidase inhibited bacterial killing in IFNLR1-/- neutrophils. B. pertussis-induced interferon lambda secretion and IFNLR1 gene expression in mouse and human neutrophils and this was dependent on the bacterial virulence protein pertussis toxin. Pertussis toxin enhanced bacterial loads in wild type but not in MRP8creIFNLR1fl/fl or IFNLR1-/- mice. Thus, pertussis toxin disrupts neutrophil function by enhancing type III IFN signaling, which prevents neutrophils from effectively clearing B. pertussis during infection, leading to higher bacterial loads and exacerbation of lung inflammation.
Collapse
Affiliation(s)
- Amit Kumar
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Da’Kuawn Johnson
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Alicia Bukowski
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Michael J. Noto
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nicholas H. Carbonetti
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
3
|
O’Neill E, Curham L, Ní Chasaide C, O’Brien S, McManus G, Moran B, Rubin K, Glazer S, Lynch MA, Mills KH. Neonatal infection with Bordetella pertussis promotes autism-like phenotypes in mice. iScience 2025; 28:111548. [PMID: 39897939 PMCID: PMC11784780 DOI: 10.1016/j.isci.2024.111548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 09/09/2024] [Accepted: 12/04/2024] [Indexed: 02/04/2025] Open
Abstract
Autism spectrum disorder (ASD) has been linked with infections early in life. Here we demonstrate that the infection of neonatal mice with the respiratory pathogen Bordetella pertussis leads to neuroinflammation, neurodevelopmental defects, and ASD-like behaviors. Following the respiratory challenge of neonatal mice with B. pertussis, multiple atypical CNS findings were observed, including blood-brain barrier disruption, dissemination of live B. pertussis bacteria to the brain with the concomitant infiltration of inflammatory monocytes, neutrophils, and activated IL-17A- and IFN-γ-producing CD4 T cells. Microglia from infected mice were activated, with impaired phagocytic function, resulting in defective synaptic pruning and disrupted neuronal circuit formation. Impaired neurodevelopment in B. pertussis-infected post-natal mice was associated with ASD-like behavioral abnormalities in young adulthood. Our data indicate that infection with virulent B. pertussis during infancy increases the risk of autism-like behavior in young adult mice. A study into the potential role of B. pertussis in human ASD is warranted.
Collapse
Affiliation(s)
- Eoin O’Neill
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
- Trinity College Institute of Neuroscience, Trinity College Dublin, D02PD91 Dublin, Ireland
| | - Lucy Curham
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| | - Caitlín Ní Chasaide
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| | - Síofra O’Brien
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| | - Gavin McManus
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| | - Barry Moran
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| | - Keith Rubin
- ILiAD Biotechnologies, Weston, FL 33331, USA
| | | | - Marina A. Lynch
- Trinity College Institute of Neuroscience, Trinity College Dublin, D02PD91 Dublin, Ireland
| | - Kingston H.G. Mills
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02R590 Dublin, Ireland
| |
Collapse
|
4
|
Mitchell AE, Scanlon KM, Flowers EM, Jordan CM, Tibbs EJ, Bukowski A, Gallop D, Carbonetti NH. Age-dependent natural killer cell and interferon γ deficits contribute to severe pertussis in infant mice. J Leukoc Biol 2024; 115:1143-1153. [PMID: 38285898 PMCID: PMC11135619 DOI: 10.1093/jleuko/qiae020] [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: 07/26/2023] [Revised: 11/29/2023] [Accepted: 12/26/2023] [Indexed: 01/31/2024] Open
Abstract
Many respiratory infections are selectively injurious to infants, yet the etiology of age-associated susceptibility is unknown. One such bacterial pathogen is Bordetella pertussis. In adult mice, innate interferon γ (IFN-γ) is produced by natural killer (NK) cells and restricts infection to the respiratory tract. In contrast, infant pertussis resembles disease in NK cell- and IFN-γ-deficient adult mice that experience disseminated lethal infection. We hypothesized that infants exhibit age-associated deficits in NK cell frequency, maturation, and responsiveness to B. pertussis, associated with low IFN-γ levels. To delineate mechanisms behind age-dependent susceptibility, we compared infant and adult mouse models of infection. Infection in infant mice resulted in impaired upregulation of IFN-γ and substantial bacterial dissemination. B. pertussis-infected infant mice displayed fewer pulmonary NK cells than adult mice. Furthermore, the NK cells in the infant mouse lungs had an immature phenotype, and the infant lung showed no upregulation of the IFN-γ-inducing cytokine IL-12p70. Adoptive transfer of adult NK cells into infants, or treatment with exogenous IFN-γ, significantly reduced bacterial dissemination. These data indicate that the lack of NK cell-produced IFN-γ significantly contributes to infant fulminant pertussis and could be the basis for other pathogen-induced, age-dependent respiratory diseases.
Collapse
Affiliation(s)
- Ashley E. Mitchell
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Karen M. Scanlon
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Emily M. Flowers
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Cassandra M. Jordan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Ellis J. Tibbs
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Alicia Bukowski
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Danisha Gallop
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Nicholas H. Carbonetti
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
5
|
Kang KR, Kim JA, Cho GW, Kang HU, Kang HM, Kang JH, Seong BL, Lee SY. Comparative Evaluation of Recombinant and Acellular Pertussis Vaccines in a Murine Model. Vaccines (Basel) 2024; 12:108. [PMID: 38276680 PMCID: PMC10818713 DOI: 10.3390/vaccines12010108] [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: 11/20/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
Since the 2000s, sporadic outbreaks of whooping cough have been reported in advanced countries, where the acellular pertussis vaccination rate is relatively high, and in developing countries. Small-scale whooping cough has also continued in many countries, due in part to the waning of immune protection after childhood vaccination, necessitating the development of an improved pertussis vaccine and vaccination program. Currently, two different production platforms are being actively pursued in Korea; one is based on the aP (acellular pertussis) vaccine purified from B. pertussis containing pertussis toxoid (PT), filamentous hemagglutin (FHA) and pertactin (PRN), and the other is based on the recombinant aP (raP), containing genetically detoxified pertussis toxin ADP-ribosyltransferase subunit 1 (PtxS1), FHA, and PRN domain, expressed and purified from recombinant E. coli. aP components were further combined with diphtheria and tetanus vaccine components as a prototype DTaP vaccine by GC Pharma (GC DTaP vaccine). We evaluated and compared the immunogenicity and the protective efficacy of aP and raP vaccines in an experimental murine challenge model: humoral immunity in serum, IgA secretion in nasal lavage, bacterial clearance after challenge, PTx (pertussis toxin) CHO cell neutralization titer, cytokine secretion in spleen single cell, and tissue resident memory CD4+ T cell (CD4+ TRM cell) in lung tissues. In humoral immunogenicity, GC DTaP vaccines showed high titers for PT and PRN and showed similar patterns in nasal lavage and IL-5 cytokine secretions. The GC DTaP vaccine and the control vaccine showed equivalent results in bacterial clearance after challenge, PTx CHO cell neutralization assay, and CD4+ TRM cell. In contrast, the recombinant raP vaccine exhibited strong antibody responses for FHA and PRN, albeit with low antibody level of PT and low titer in PTx CHO neutralization assay, as compared to control and GC DTaP vaccines. The raP vaccine provided a sterile lung bacterial clearance comparable to a commercial control vaccine after the experimental challenge in murine model. Moreover, raP exhibited a strong cytokine response and CD4+ TRM cell in lung tissue, comparable or superior to the experimental and commercial DTaP vaccinated groups. Contingent on improving the biophysical stability and humoral response to PT, the raP vaccine warrants further development as an effective alternative to aP vaccines for the control of a pertussis outbreak.
Collapse
Affiliation(s)
- Kyu-Ri Kang
- The Vaccine Bio Research Institute, Annex to Seoul Saint Mary Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea (J.-H.K.)
| | - Ji-Ahn Kim
- The Vaccine Bio Research Institute, Annex to Seoul Saint Mary Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea (J.-H.K.)
| | - Gyu-Won Cho
- The Vaccine Bio Research Institute, Annex to Seoul Saint Mary Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea (J.-H.K.)
| | - Han-Ul Kang
- The Interdisciplinary Graduate Program in Integrative Biotechnology, Yonsei University, Incheon 21983, Republic of Korea
| | - Hyun-Mi Kang
- The Vaccine Bio Research Institute, Annex to Seoul Saint Mary Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea (J.-H.K.)
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jin-Han Kang
- The Vaccine Bio Research Institute, Annex to Seoul Saint Mary Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea (J.-H.K.)
| | - Baik-Lin Seong
- Department of Microbiology and Immunology, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| | - Soo-Young Lee
- The Vaccine Bio Research Institute, Annex to Seoul Saint Mary Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea (J.-H.K.)
- Department of Pediatrics, Bucheon St. Mary’s Hospital, The Catholic University of Korea, Bucheon 14647, Republic of Korea
| |
Collapse
|
6
|
Abu-Raya B, Esser MJ, Nakabembe E, Reiné J, Amaral K, Diks AM, Imede E, Way SS, Harandi AM, Gorringe A, Le Doare K, Halperin SA, Berkowska MA, Sadarangani M. Antibody and B-cell Immune Responses Against Bordetella Pertussis Following Infection and Immunization. J Mol Biol 2023; 435:168344. [PMID: 37926426 DOI: 10.1016/j.jmb.2023.168344] [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: 08/30/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
Neither immunization nor recovery from natural infection provides life-long protection against Bordetella pertussis. Replacement of a whole-cell pertussis (wP) vaccine with an acellular pertussis (aP) vaccine, mutations in B. pertussis strains, and better diagnostic techniques, contribute to resurgence of number of cases especially in young infants. Development of new immunization strategies relies on a comprehensive understanding of immune system responses to infection and immunization and how triggering these immune components would ensure protective immunity. In this review, we assess how B cells, and their secretory products, antibodies, respond to B. pertussis infection, current and novel vaccines and highlight similarities and differences in these responses. We first focus on antibody-mediated immunity. We discuss antibody (sub)classes, elaborate on antibody avidity, ability to neutralize pertussis toxin, and summarize different effector functions, i.e. ability to activate complement, promote phagocytosis and activate NK cells. We then discuss challenges and opportunities in studying B-cell immunity. We highlight shared and unique aspects of B-cell and plasma cell responses to infection and immunization, and discuss how responses to novel immunization strategies better resemble those triggered by a natural infection (i.e., by triggering responses in mucosa and production of IgA). With this comprehensive review, we aim to shed some new light on the role of B cells and antibodies in the pertussis immunity to guide new vaccine development.
Collapse
Affiliation(s)
- Bahaa Abu-Raya
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada.
| | - Mirjam J Esser
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Eve Nakabembe
- Centre for Neonatal and Paediatric Infectious Diseases Research, St George's, University of London, Cranmer Terrace, London SW17 0RE, UK; Department of Obstetrics and Gynaecology, Makerere University College of Health Sciences, Upper Mulago Hill Road, Kampala, P.O. Box 7072, Uganda
| | - Jesús Reiné
- Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom; Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
| | - Kyle Amaral
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Annieck M Diks
- Department of Immunology, Leiden University Medical Center, Albinusdreef 2, Leiden ZA 2333, the Netherlands
| | - Esther Imede
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | - Sing Sing Way
- Department of Pediatrics, Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Ali M Harandi
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Andrew Gorringe
- UK Health Security Agency, Porton Down, Salisbury SP4 0JG, UK
| | - Kirsty Le Doare
- Centre for Neonatal and Paediatric Infectious Diseases Research, St George's, University of London, Cranmer Terrace, London SW17 0RE, UK; Makerere University-Johns Hopkins University Research Collaboration, MU-JHU, Upper Mulago Hill, Kampala, P.O. Box 23491, Uganda
| | - Scott A Halperin
- Canadian Center for Vaccinology, Departments of Pediatrics and Microbiology and Immunology, Dalhousie University, Izaak Walton Killam Health Centre, and Nova Scotia Health Authority, Halifax, NS, Canada
| | - Magdalena A Berkowska
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Manish Sadarangani
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
7
|
Weaver KL, Bitzer GJ, Wolf MA, Pyles GM, DeJong MA, Dublin SR, Huckaby AB, Gutierrez MDLP, Hall JM, Wong TY, Warden M, Petty JE, Witt WT, Cunningham C, Sen-Kilic E, Damron FH, Barbier M. Intranasal challenge with B. pertussis leads to more severe disease manifestations in mice than aerosol challenge. PLoS One 2023; 18:e0286925. [PMID: 37917623 PMCID: PMC10621807 DOI: 10.1371/journal.pone.0286925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/25/2023] [Indexed: 11/04/2023] Open
Abstract
The murine Bordetella pertussis challenge model has been utilized in preclinical research for decades. Currently, inconsistent methodologies are employed by researchers across the globe, making it difficult to compare findings. The objective of this work was to utilize the CD-1 mouse model with two routes of challenge, intranasal and aerosol administration of B. pertussis, to understand the differences in disease manifestation elicited via each route. We observed that both routes of B. pertussis challenge result in dose-dependent colonization of the respiratory tract, but overall, intranasal challenge led to higher bacterial burden in the nasal lavage, trachea, and lung. Furthermore, high dose intranasal challenge results in induction of leukocytosis and pro-inflammatory cytokine responses compared to aerosol challenge. These data highlight crucial differences in B. pertussis challenge routes that should be considered during experimental design.
Collapse
Affiliation(s)
- Kelly L. Weaver
- Vaccine Development Center in the Department of Microbiology, Immunology, and Cell Biology at West Virginia University, Morgantown, WV, United States of America
| | - Graham J. Bitzer
- Vaccine Development Center in the Department of Microbiology, Immunology, and Cell Biology at West Virginia University, Morgantown, WV, United States of America
| | - M. Allison Wolf
- Vaccine Development Center in the Department of Microbiology, Immunology, and Cell Biology at West Virginia University, Morgantown, WV, United States of America
| | - Gage M. Pyles
- Vaccine Development Center in the Department of Microbiology, Immunology, and Cell Biology at West Virginia University, Morgantown, WV, United States of America
| | - Megan A. DeJong
- Vaccine Development Center in the Department of Microbiology, Immunology, and Cell Biology at West Virginia University, Morgantown, WV, United States of America
| | - Spencer R. Dublin
- Vaccine Development Center in the Department of Microbiology, Immunology, and Cell Biology at West Virginia University, Morgantown, WV, United States of America
| | - Annalisa B. Huckaby
- Vaccine Development Center in the Department of Microbiology, Immunology, and Cell Biology at West Virginia University, Morgantown, WV, United States of America
| | - Maria de la Paz Gutierrez
- Vaccine Development Center in the Department of Microbiology, Immunology, and Cell Biology at West Virginia University, Morgantown, WV, United States of America
| | - Jesse M. Hall
- Vaccine Development Center in the Department of Microbiology, Immunology, and Cell Biology at West Virginia University, Morgantown, WV, United States of America
| | - Ting Y. Wong
- Vaccine Development Center in the Department of Microbiology, Immunology, and Cell Biology at West Virginia University, Morgantown, WV, United States of America
| | - Matthew Warden
- Vaccine Development Center in the Department of Microbiology, Immunology, and Cell Biology at West Virginia University, Morgantown, WV, United States of America
| | - Jonathan E. Petty
- Vaccine Development Center in the Department of Microbiology, Immunology, and Cell Biology at West Virginia University, Morgantown, WV, United States of America
| | - William T. Witt
- Vaccine Development Center in the Department of Microbiology, Immunology, and Cell Biology at West Virginia University, Morgantown, WV, United States of America
| | - Casey Cunningham
- Vaccine Development Center in the Department of Microbiology, Immunology, and Cell Biology at West Virginia University, Morgantown, WV, United States of America
| | - Emel Sen-Kilic
- Vaccine Development Center in the Department of Microbiology, Immunology, and Cell Biology at West Virginia University, Morgantown, WV, United States of America
| | - F. Heath Damron
- Vaccine Development Center in the Department of Microbiology, Immunology, and Cell Biology at West Virginia University, Morgantown, WV, United States of America
| | - Mariette Barbier
- Vaccine Development Center in the Department of Microbiology, Immunology, and Cell Biology at West Virginia University, Morgantown, WV, United States of America
| |
Collapse
|
8
|
Schmitt P, Borkner L, Jazayeri SD, McCarthy KN, Mills KH. Nasal vaccines for pertussis. Curr Opin Immunol 2023; 84:102355. [PMID: 37307651 DOI: 10.1016/j.coi.2023.102355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 06/14/2023]
Abstract
Whooping cough, caused by Bordetella pertussis, is still a major cause of morbidity and mortality worldwide. Current acellular pertussis (aP) vaccines induce potent circulating IgG and prevent severe disease in children/adults and in infants born to vaccinated mothers. However, they do not prevent nasal infection, allowing asymptomatic transmission of B. pertussis. Studies in animal models have demonstrated that, unlike natural infection, immunization with aP vaccines fails to induce secretory immunoglobulin A (IgA) or interleukin-17 (IL-17)-secreting tissue-resident memory CD4 T (TRM) cells, required for sustained sterilizing immunity in the nasal mucosa. Live-attenuated vaccines or aP vaccines formulated with novel adjuvants that induce respiratory IgA and TRM cells, especially when delivered by the nasal route, are in development and have considerable promise as next-generation vaccines against pertussis.
Collapse
Affiliation(s)
- Pauline Schmitt
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Lisa Borkner
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Seyed Davoud Jazayeri
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Karen N McCarthy
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Kingston Hg Mills
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
9
|
Miguelena Chamorro B, De Luca K, Swaminathan G, Longet S, Mundt E, Paul S. Bordetella bronchiseptica and Bordetella pertussis: Similarities and Differences in Infection, Immuno-Modulation, and Vaccine Considerations. Clin Microbiol Rev 2023; 36:e0016422. [PMID: 37306571 PMCID: PMC10512794 DOI: 10.1128/cmr.00164-22] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023] Open
Abstract
Bordetella pertussis and Bordetella bronchiseptica belong to the genus Bordetella, which comprises 14 other species. B. pertussis is responsible for whooping cough in humans, a severe infection in children and less severe or chronic in adults. These infections are restricted to humans and currently increasing worldwide. B. bronchiseptica is involved in diverse respiratory infections in a wide range of mammals. For instance, the canine infectious respiratory disease complex (CIRDC), characterized by a chronic cough in dogs. At the same time, it is increasingly implicated in human infections, while remaining an important pathogen in the veterinary field. Both Bordetella can evade and modulate host immune responses to support their persistence, although it is more pronounced in B. bronchiseptica infection. The protective immune responses elicited by both pathogens are comparable, while there are important characteristics in the mechanisms that differ. However, B. pertussis pathogenesis is more difficult to decipher in animal models than those of B. bronchiseptica because of its restriction to humans. Nevertheless, the licensed vaccines for each Bordetella are different in terms of formulation, route of administration and immune responses induced, with no known cross-reaction between them. Moreover, the target of the mucosal tissues and the induction of long-lasting cellular and humoral responses are required to control and eliminate Bordetella. In addition, the interaction between both veterinary and human fields are essential for the control of this genus, by preventing the infections in animals and the subsequent zoonotic transmission to humans.
Collapse
Affiliation(s)
- Beatriz Miguelena Chamorro
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, Lyon, France
- Boehringer Ingelheim, Global Innovation, Saint-Priest, France
| | - Karelle De Luca
- Boehringer Ingelheim, Global Innovation, Saint-Priest, France
| | | | - Stéphanie Longet
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, Lyon, France
- CIC Inserm 1408 Vaccinology, Saint-Etienne, France
| | - Egbert Mundt
- Boehringer Ingelheim, Global Innovation, Saint-Priest, France
| | - Stéphane Paul
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP (Saint-Etienne), Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, UJM, Lyon, France
- CIC Inserm 1408 Vaccinology, Saint-Etienne, France
| |
Collapse
|
10
|
Yang B, Zhu D, Zhou Y, Gong B, Hu Y, Zhang J, Huang S, Nian X, Li X, Li X, Duan K, Yang X. Liposome and QS-21 Combined Adjuvant Induces theHumoral and Cellular Responses of Acellular Pertussis Vaccine in a Mice Model. Vaccines (Basel) 2023; 11:vaccines11050914. [PMID: 37243018 DOI: 10.3390/vaccines11050914] [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: 03/15/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
The resurgence of pertussis in vaccinated communities may be related to the reduced long-term immunity induced by acellular pertussis vaccines. Therefore, developing improved pertussis vaccine candidates that could induce strong Th1 or Th17 cellular immunity is an urgent need. The use of new adjuvants may well meet this requirement. In this research, we developed a novel adjuvant candidate by combining liposome and QS-21 adjuvant. Adjuvant activity, protective efficacy, the level of neutralizing antibody against PT, and the resident memory T (TRM) cells in lung tissue after vaccination were studied. We then performed B. pertussis respiratory challenge in mice after they received vaccination with traditional aluminum hydroxide and the novel adjuvant combination. Results showed that the liposome + QS-21 adjuvant group had a rapid antibody and higher antibody (PT, FHA, Fim) level, induced anti-PT neutralizing antibody and recruited more IL-17A-secreting CD4+ TRM cells along with IL-17A-secreting CD8+ TRM cells in mice, which provided robust protection against B. pertussis infection. These results provide a key basis for liposome + QS-21 adjuvant as a promising adjuvant candidate for developing an acellular pertussis vaccine that elicits protective immunity against pertussis.
Collapse
Affiliation(s)
- Baifeng Yang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Dewu Zhu
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Yisi Zhou
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Beizhe Gong
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Yuan Hu
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Jiayou Zhang
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Shihe Huang
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Xuanxuan Nian
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Xinghang Li
- National Engineering Technology Research Center for Combined Vaccines, Wuhan 430207, China
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Xinguo Li
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Kai Duan
- Wuhan Institute of Biological Products Co., Ltd., Wuhan 430207, China
| | - Xiaoming Yang
- National Biotec Group Company Limited, Beijing 100024, China
| |
Collapse
|
11
|
Szwejser-Zawislak E, Wilk MM, Piszczek P, Krawczyk J, Wilczyńska D, Hozbor D. Evaluation of Whole-Cell and Acellular Pertussis Vaccines in the Context of Long-Term Herd Immunity. Vaccines (Basel) 2022; 11:vaccines11010001. [PMID: 36679846 PMCID: PMC9863224 DOI: 10.3390/vaccines11010001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/04/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
After the pertussis vaccine had been introduced in the 1940s and was shown to be very successful in reducing the morbidity and mortality associated with the disease, the possibility of improving both vaccine composition and vaccination schedules has become the subject of continuous interest. As a result, we are witnessing a considerable heterogeneity in pertussis vaccination policies, which remains beyond universal consensus. Many pertussis-related deaths still occur in low- and middle-income countries; however, these deaths are attributable to gaps in vaccination coverage and limited access to healthcare in these countries, rather than to the poor efficacy of the first generation of pertussis vaccine consisting in inactivated and detoxified whole cell pathogen (wP). In many, particularly high-income countries, a switch was made in the 1990s to the use of acellular pertussis (aP) vaccine, to reduce the rate of post-vaccination adverse events and thereby achieve a higher percentage of children vaccinated. However the epidemiological data collected over the past few decades, even in those high-income countries, show an increase in pertussis prevalence and morbidity rates, triggering a wide-ranging debate on the causes of pertussis resurgence and the effectiveness of current pertussis prevention strategies, as well as on the efficacy of available pertussis vaccines and immunization schedules. The current article presents a systematic review of scientific reports on the evaluation of the use of whole-cell and acellular pertussis vaccines, in the context of long-term immunity and vaccines efficacy.
Collapse
Affiliation(s)
- Ewa Szwejser-Zawislak
- Institute of Biotechnology of Serums and Vaccines Biomed, Al. Sosnowa 8, 30-224 Krakow, Poland
| | - Mieszko M. Wilk
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | - Piotr Piszczek
- Institute of Biotechnology of Serums and Vaccines Biomed, Al. Sosnowa 8, 30-224 Krakow, Poland
| | - Justyna Krawczyk
- Institute of Biotechnology of Serums and Vaccines Biomed, Al. Sosnowa 8, 30-224 Krakow, Poland
| | - Daria Wilczyńska
- Institute of Biotechnology of Serums and Vaccines Biomed, Al. Sosnowa 8, 30-224 Krakow, Poland
| | - Daniela Hozbor
- VacSal Laboratory, Institute of Biotechnology and Molecular Biology, Faculty of Sciences, National University of La Plata (UNLP), National Council for Scientific and Technical Research (CONICET), La Plata 1900, Argentina
- Correspondence:
| |
Collapse
|
12
|
Jiang W, Wang X, Su Y, Cai L, Li J, Liang J, Gu Q, Sun M, Shi L. Intranasal Immunization With a c-di-GMP-Adjuvanted Acellular Pertussis Vaccine Provides Superior Immunity Against Bordetella pertussis in a Mouse Model. Front Immunol 2022; 13:878832. [PMID: 35493458 PMCID: PMC9043693 DOI: 10.3389/fimmu.2022.878832] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/22/2022] [Indexed: 12/03/2022] Open
Abstract
Pertussis, caused by the gram-negative bacterium Bordetella pertussis, is a highly contagious respiratory disease. Intranasal vaccination is an ideal strategy to prevent pertussis, as the nasal mucosa represents the first-line barrier to B. pertussis infection. The current intramuscular acellular pertussis (aP) vaccines elicit strong antibody and Th2-biased responses but not necessary cellular and mucosal immunity. Here, we formulated two cyclic dinucleotide (CDN)-adjuvanted aP subunit vaccines, a mammalian 2’,3’-cGAMP-adjuvanted aP vaccine and a bacterial-derived c-di-GMP-adjuvanted aP vaccine, and evaluated their immunogenicity in a mouse model. We found that the aP vaccine alone delivered intranasally (IN) induced moderate systemic and mucosal humoral immunity but weak cellular immunity, whereas the alum-adjuvanted aP vaccine administered intraperitoneally elicited higher Th2 and systemic humoral immune responses but weaker Th1 and Th17 and mucosal immune responses. In contrast, both CDN-adjuvanted aP vaccines administered via the IN route induced robust humoral and cellular immunity systemically and mucosally. Furthermore, the c-di-GMP-adjuvanted aP vaccine generated better antibody production and stronger Th1 and Th17 responses than the 2′,3′-cGAMP-adjuvanted aP vaccine. In addition, following B. pertussis challenge, the group of mice that received IN immunization with the c-di-GMP-adjuvanted aP vaccine showed better protection than all other groups of vaccinated mice, with decreased inflammatory cell infiltration in the lung and reduced bacterial burden in both the upper and lower respiratory tracts. In summary, the c-di-GMP-adjuvanted aP vaccine can elicit a multifaceted potent immune response resulting in robust bacterial clearance in the respiratory tract, which indicates that c-di-GMP can serve as a potential mucosal adjuvant for the pertussis vaccine.
Collapse
Affiliation(s)
- Wenwen Jiang
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China
| | - Xiaoyu Wang
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China
| | - Yuhao Su
- Laboratory of Immunogenetics, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China
| | - Lukui Cai
- Laboratory of Vaccine Development, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China
| | - Jingyan Li
- Laboratory of Vaccine Development, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China
| | - Jiangli Liang
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China
| | - Qin Gu
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China
| | - Mingbo Sun
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China.,Laboratory of Vaccine Development, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China
| | - Li Shi
- Laboratory of Immunogenetics, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming, China
| |
Collapse
|
13
|
Lambert EE, van Twillert I, Beckers L, Poelen MCM, Han WGH, Pieren DKJ, van Els CACM. Reduced Bordetella pertussis-specific CD4+ T-Cell Responses at Older Age. FRONTIERS IN AGING 2022; 2:737870. [PMID: 35822011 PMCID: PMC9261443 DOI: 10.3389/fragi.2021.737870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022]
Abstract
Pertussis, a human-specific respiratory infectious disease caused by the Gram-negative bacterium Bordetella pertussis (Bp), remains endemic with epidemic years despite high vaccination coverage. Whereas pertussis vaccines and natural infection with Bp confer immune protection, the duration of protection varies and is not lifelong. Recent evidence indicates a considerable underestimation of the pertussis burden among older adults. Whereas the impact of increasing age on Bp-specific humoral immunity has been demonstrated, little is known on immunosenescence of CD4+ T-cell responses in the context of Bp. Here, we aimed to address whether increasing age impacts responsiveness of the Bp-specific CD4+ T-cells in the memory pool following a clinically symptomatic pertussis infection in whole cell vaccine-primed pediatric and adult cases. Cytokine and proliferative responses and phenotypical profiles of CD4+ T cells specific for Bp antigens at an early and late convalescent timepoint were compared. Responses of various Th cytokines, including IFNγ, were significantly lower in older adults at early and late timepoints post diagnosis. In addition, we found lower frequencies of Bp-specific proliferated CD4+ T cells in older adults, in the absence of differences in replication profile. Phenotyping of Bp-specific CD4+ T cells suggested reduced expression of activation markers rather than increased expression of co-inhibitory markers. Altogether, our findings show that the magnitude and functionality of the Bp-specific memory CD4+ T-cell pool decrease at older age. Declined CD4+ T-cell responsiveness to Bp is suggested to contribute to the burden of pertussis in older adults.
Collapse
|
14
|
Cimolai N. Non-primate animal models for pertussis: back to the drawing board? Appl Microbiol Biotechnol 2022; 106:1383-1398. [PMID: 35103810 PMCID: PMC8803574 DOI: 10.1007/s00253-022-11798-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 11/30/2022]
Abstract
Despite considerable progress in the understanding of clinical pertussis, the contemporary emergence of antimicrobial resistance for Bordetella pertussis and an evolution of concerns with acellular component vaccination have both sparked a renewed interest. Although simian models of infection best correlate with the observed attributes of human infection, several animal models have been used for decades and have positively contributed in many ways to the related science. Nevertheless, there is yet the lack of a reliable small animal model system that mimics the combination of infection genesis, variable upper and lower respiratory infection, systemic effects, infection resolution, and vaccine responses. This narrative review examines the history and attributes of non-primate animal models for pertussis and places context with the current use and needs. Emerging from the latter is the necessity for further such study to better create the optimal model of infection and vaccination with use of current molecular tools and a broader range of animal systems. KEY POINTS: • Currently used and past non-primate animal models of B. pertussis infection often have unique and focused applications. • A non-primate animal model that consistently mimics human pertussis for the majority of key infection characteristics is lacking. • There remains ample opportunity for an improved non-primate animal model of pertussis with the use of current molecular biology tools and with further exploration of species not previously considered.
Collapse
Affiliation(s)
- Nevio Cimolai
- Faculty of Medicine, The University of British Columbia, Vancouver, Canada.
- Children's and Women's Health Centre of British Columbia, 4480 Oak Street, Vancouver, B.C., V6H3V4, Canada.
| |
Collapse
|
15
|
Abstract
The Bordetella pertussis bacterium is the causative agent of whooping cough (pertussis disease). Following recent outbreaks of pertussis, disproportionately affecting young infants, several countries have introduced maternal pertussis vaccination strategies, aimed at boosting transplacental transfer of protective antibodies during pregnancy. Given historical associations between high maternal antibody and blunted infant responses to vaccination, subsequent research studies have investigated the impact of maternal pertussis vaccine on infant humoral responses. However, far less is known about the potential impact of the vaccine on innate immunity. Here, we describe methods to detect in vitro cellular responses to B. pertussis in mothers and their infants using a B. pertussis stimulation assay and multiplex cytokine assays to address this research question.
Collapse
|
16
|
Mucosal Immunization with DTaP Confers Protection against Bordetella pertussis Infection and Cough in Sprague-Dawley Rats. Infect Immun 2021; 89:e0034621. [PMID: 34516235 PMCID: PMC8594602 DOI: 10.1128/iai.00346-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Pertussis is a respiratory disease caused by the Gram-negative pathogen, Bordetella pertussis. The transition from a whole-cell pertussis vaccine (wP and DTP) to an acellular pertussis vaccine (aP, DTaP, and Tdap) correlates with an increase in pertussis cases, despite widespread vaccine implementation and coverage, and it is now appreciated that the protection provided by aP rapidly wanes. To recapitulate the localized immunity observed from natural infection, mucosal vaccination with aP was explored using the coughing rat model of pertussis. Overall, our goal was to evaluate the route of vaccination in the coughing rat model of pertussis. Immunity induced by both oral gavage and intranasal vaccination of aP in B. pertussis challenged rats over a 9-day infection was compared to intramuscular wP (IM-wP)- and IM-aP-immunized rats that were used as positive controls. Our data demonstrate that mucosal immunization of aP resulted in the production of anti-B. pertussis IgG antibody titers similar to IM-wP- and IM-aP-vaccinated controls postchallenge. IN-aP also induced anti-B. pertussis IgA antibodies in the nasal cavity. Immunization with IM-wP, IM-aP, IN-aP, and OG-aP immunization protected against B. pertussis-induced cough, whereas OG-aP immunization did not protect against respiratory distress. Mucosal immunization by both intranasal and oral gavage administration protected against acute inflammation and decreased bacterial burden in the lung compared to mock-vaccinated challenge rats. The data presented in this study suggest that mucosal vaccination with aP can induce a mucosal immune response and provide protection against B. pertussis challenge. This study highlights the potential benefits and uses of the coughing rat model of pertussis; however, further questions regarding waning immunity still require additional investigation.
Collapse
|
17
|
Kang KR, Huh DH, Kim JA, Kang JH. Immunogenicity of a new enhanced tetanus-reduced dose diphtheria-acellular pertussis (Tdap) vaccine against Bordetella pertussis in a murine model. BMC Immunol 2021; 22:68. [PMID: 34641798 PMCID: PMC8506493 DOI: 10.1186/s12865-021-00457-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Background The necessity of the tetanus-reduced dose diphtheria-acellular pertussis (Tdap) vaccine in adolescence and adults has been emphasized since the resurgence of small-scale pertussis in Korea and worldwide due to the waning effect of the vaccine and variant pathogenic stains in the late 1990s. GreenCross Pharma (GC Pharma), a Korean company, developed the Tdap vaccine GC3111 in 2010. Recently, they enhanced the vaccine, GC3111, produced previously in 2010 to reinforce the antibody response against filamentous hemagglutinin (FHA). In this study, immunogenicity and efficacy of the enhanced Tdap vaccine compared and evaluated with two Tdap vaccines, GC3111 vaccine produced in 2010 previously and commercially available Tdap vaccine in a murine model. Methods Two tests groups and positive control group of Balb/c mice were primed with two doses of the diphtheria-tetanus-acellular pertussis (DTaP) vaccine followed by a single booster Tdap vaccine at 9 week using the commercially available Tdap vaccine or 2 Tdap vaccines from GC Pharma (GC3111, enhanced GC3111). Humoral response was assessed 1 week before and 2 and 4 weeks after Tdap booster vaccination. The enhanced GC3111 generated similar humoral response compare to the commercial vaccine for filamentous hemagglutinin (FHA). The interferon gamma (IFN-γ) (Th1), interleukin 5 (IL-5) (Th2) and interleukin 17 (IL-17) (Th17) cytokines were assessed 4 weeks after booster vaccination by stimulation with three simulators: heat inactivated Bordetella pertussis (hBp), vaccine antigens, and hBp mixed with antigens (hBp + antigen). A bacterial challenge test was performed 4 weeks after booster vaccination. Results Regarding cell-mediated immunity, cytokine secretion differed among the three simulators. However, no difference was found between two test groups and positive control group. All the vaccinated groups indicated a Th1 or Th1/Th2 response. On Day 5 post-bacterial challenge, B. pertussis colonies were absent in the lungs in two test groups and positive control group. Conclusions Our results confirmed the immunogenicity of GC Pharma’s Tdap vaccine; enhanced GC3111 was equivalent to the presently used commercial vaccine in terms of humoral response as well as cell-mediated cytokine expression. Supplementary Information The online version contains supplementary material available at 10.1186/s12865-021-00457-1.
Collapse
Affiliation(s)
- Kyu Ri Kang
- The Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Annex to Seoul Saint Mary Hospital, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Korea
| | - Dong Ho Huh
- The Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Annex to Seoul Saint Mary Hospital, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Korea
| | - Ji Ahn Kim
- The Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Annex to Seoul Saint Mary Hospital, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Korea
| | - Jin Han Kang
- The Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Annex to Seoul Saint Mary Hospital, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Korea. .,Department of Pediatrics, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Korea.
| |
Collapse
|
18
|
Rice TF, Diavatopoulos DA, Guo Y, Donaldson B, Bouqueau M, Bosanquet A, Barnett S, Holder B, Kampmann B. Modification of innate immune responses to Bordetella pertussis in babies from pertussis vaccinated pregnancies. EBioMedicine 2021; 72:103612. [PMID: 34649076 PMCID: PMC8517834 DOI: 10.1016/j.ebiom.2021.103612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/14/2021] [Accepted: 09/17/2021] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Tetanus, diphtheria, acellular pertussis, inactivated polio (Tdap-IPV) vaccines administered during pregnancy protect young infants from Bordetella pertussis (B. pertussis) infection. Whilst the impact of maternal Tdap-IPV vaccination on infants' humoral response to subsequent pertussis immunisation has been investigated, little is known about any impact on innate responses. METHODS We investigated the immune response to B. pertussis in mothers and infants from Tdap-IPV-vaccinated and unvaccinated pregnancies, utilising a whole blood assay and flow cytometric phenotyping of neonatal natural killer (NK) cells, monocytes and dendritic cells. Blood was collected from mother and umbilical cord at birth, and from infants at seven weeks (one week pre-primary pertussis immunisation) and five months of age (one month post-primary pertussis immunisation). 21 mothers and 67 infants were studied. FINDINGS Vaccinated women had elevated pro-inflammatory cytokine responses to B. pertussis. At birth, babies of vaccinated women had elevated IL-2 and IL-12 responses, elevated classical monocyte proportions, and reduced monocyte and NK cell cytokine responses. The elevated IL-2 response persisted to seven weeks-of-age, when lower IL-10 and IL-13 responses were also seen. One-month post-primary pertussis vaccination, infants from vaccinated pregnancies still had lower IL-10 responses to B. pertussis, as well as lower IL-4. INTERPRETATION This study suggests that pertussis vaccination during pregnancy impacts infant cellular immune responses, potentially contributing to the modification of antibody responses already reported following primary immunisation against B. pertussis. FUNDING National Institute for Health Research Imperial Biomedical Research Centre and IMmunising PRegnant women and INfants neTwork (funded by the GCRF Networks in Vaccines R&D).
Collapse
Affiliation(s)
- Thomas F Rice
- Department of Metabolism, Development and Reproduction (MDR), Lecturer in Maternal and Fetal Health, Imperial College London, Institute of Reproductive and Developmental Biology (IRDB), Hammersmith Campus, London W12 0HS, UK; Section of Paediatrics, Department of Medicine, Imperial College London, UK
| | - Dimitri A Diavatopoulos
- Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Yanping Guo
- National Heart and Lung Institute (NHLI), Imperial College London, UK
| | - Beverly Donaldson
- Section of Paediatrics, Department of Medicine, Imperial College London, UK
| | - Marielle Bouqueau
- Section of Paediatrics, Department of Medicine, Imperial College London, UK
| | - Anna Bosanquet
- Section of Paediatrics, Department of Medicine, Imperial College London, UK
| | - Sara Barnett
- Department of Metabolism, Development and Reproduction (MDR), Lecturer in Maternal and Fetal Health, Imperial College London, Institute of Reproductive and Developmental Biology (IRDB), Hammersmith Campus, London W12 0HS, UK
| | - Beth Holder
- Department of Metabolism, Development and Reproduction (MDR), Lecturer in Maternal and Fetal Health, Imperial College London, Institute of Reproductive and Developmental Biology (IRDB), Hammersmith Campus, London W12 0HS, UK; Section of Paediatrics, Department of Medicine, Imperial College London, UK.
| | - Beate Kampmann
- Section of Paediatrics, Department of Medicine, Imperial College London, UK; The Vaccine Centre, London School of Hygiene and Tropical Medicine, UK; Vaccines and Immunity Theme, MRC Unit The Gambia at LSHTM, Gambia
| |
Collapse
|
19
|
Saso A, Kampmann B, Roetynck S. Vaccine-Induced Cellular Immunity against Bordetella pertussis: Harnessing Lessons from Animal and Human Studies to Improve Design and Testing of Novel Pertussis Vaccines. Vaccines (Basel) 2021; 9:877. [PMID: 34452002 PMCID: PMC8402596 DOI: 10.3390/vaccines9080877] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 12/14/2022] Open
Abstract
Pertussis ('whooping cough') is a severe respiratory tract infection that primarily affects young children and unimmunised infants. Despite widespread vaccine coverage, it remains one of the least well-controlled vaccine-preventable diseases, with a recent resurgence even in highly vaccinated populations. Although the exact underlying reasons are still not clear, emerging evidence suggests that a key factor is the replacement of the whole-cell (wP) by the acellular pertussis (aP) vaccine, which is less reactogenic but may induce suboptimal and waning immunity. Differences between vaccines are hypothesised to be cell-mediated, with polarisation of Th1/Th2/Th17 responses determined by the composition of the pertussis vaccine given in infancy. Moreover, aP vaccines elicit strong antibody responses but fail to protect against nasal colonisation and/or transmission, in animal models, thereby potentially leading to inadequate herd immunity. Our review summarises current knowledge on vaccine-induced cellular immune responses, based on mucosal and systemic data collected within experimental animal and human vaccine studies. In addition, we describe key factors that may influence cell-mediated immunity and how antigen-specific responses are measured quantitatively and qualitatively, at both cellular and molecular levels. Finally, we discuss how we can harness this emerging knowledge and novel tools to inform the design and testing of the next generation of improved infant pertussis vaccines.
Collapse
Affiliation(s)
- Anja Saso
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| | - Beate Kampmann
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| | - Sophie Roetynck
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| |
Collapse
|
20
|
Dubois V, Locht C. Mucosal Immunization Against Pertussis: Lessons From the Past and Perspectives. Front Immunol 2021; 12:701285. [PMID: 34211481 PMCID: PMC8239240 DOI: 10.3389/fimmu.2021.701285] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 05/26/2021] [Indexed: 01/11/2023] Open
Abstract
Background Current vaccination strategies against pertussis are sub-optimal. Optimal protection against Bordetella pertussis, the causative agent of pertussis, likely requires mucosal immunity. Current pertussis vaccines consist of inactivated whole B. pertussis cells or purified antigens thereof, combined with diphtheria and tetanus toxoids. Although they are highly protective against severe pertussis disease, they fail to elicit mucosal immunity. Compared to natural infection, immune responses following immunization are short-lived and fail to prevent bacterial colonization of the upper respiratory tract. To overcome these shortcomings, efforts have been made for decades, and continue to be made, toward the development of mucosal vaccines against pertussis. Objectives In this review we systematically analyzed published literature on protection conferred by mucosal immunization against pertussis. Immune responses mounted by these vaccines are summarized. Method The PubMed Library database was searched for published studies on mucosal pertussis vaccines. Eligibility criteria included mucosal administration and the evaluation of at least one outcome related to efficacy, immunogenicity and safety. Results While over 349 publications were identified by the search, only 63 studies met the eligibility criteria. All eligible studies are included here. Initial attempts of mucosal whole-cell vaccine administration in humans provided promising results, but were not followed up. More recently, diverse vaccination strategies have been tested, including non-replicating and replicating vaccine candidates given by three different mucosal routes: orally, nasally or rectally. Several adjuvants and particulate formulations were tested to enhance the efficacy of non-replicating vaccines administered mucosally. Most novel vaccine candidates were only tested in animal models, mainly mice. Only one novel mucosal vaccine candidate was tested in baboons and in human trials. Conclusion Three vaccination strategies drew our attention, as they provided protective and durable immunity in the respiratory tract, including the upper respiratory tract: acellular vaccines adjuvanted with lipopeptide LP1569 and c-di-GMP, outer membrane vesicles and the live attenuated BPZE1 vaccine. Among all experimental vaccines, BPZE1 is the only one that has advanced into clinical development.
Collapse
Affiliation(s)
- Violaine Dubois
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Camille Locht
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
21
|
Triggering receptor expressed on myeloid cells-1 (TREM-1) contributes to Bordetella pertussis inflammatory pathology. Infect Immun 2021; 89:e0012621. [PMID: 34097504 DOI: 10.1128/iai.00126-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Whooping cough (pertussis) is a severe pulmonary infectious disease caused by the bacteria Bordetella pertussis. Pertussis infects an estimated 24 million people annually, resulting in >150,000 deaths. The NIH placed pertussis on the list of emerging pathogens in 2015. Antibiotics are ineffective unless administered before the onset of the disease characteristic cough. Therefore, there is an urgent need for novel pertussis therapeutics. We have shown that sphingosine-1-phosphate receptor (S1PR) agonists reduce pertussis inflammation, without increasing bacterial burden. Transcriptomic studies were performed to identify this mechanism and allow for the development of pertussis therapeutics which specifically target problematic inflammation without sacrificing bacterial control. These data suggested a role for triggering receptor expressed on myeloid cells-1 (TREM-1). TREM-1 cell surface receptor functions as an amplifier of inflammatory responses. Expression of TREM-1 is increased in response to bacterial infection of mucosal surfaces. In mice, B. pertussis infection results in TLR9-dependent increased expression of TREM-1 and its associated cytokines. Interestingly, S1PR agonists dampen pulmonary inflammation and TREM-1 expression. Mice challenged intranasally with B. pertussis and treated with ligand-dependent (LP17) and ligand-independent (GF9) TREM-1 inhibitors showed no differences in bacterial burden and significantly reduced TNF-α and CCL-2 expression compared to controls. Mice receiving TREM-1 inhibitors showed reduced pulmonary inflammation compared to controls indicating that TREM-1 promotes inflammatory pathology, but not bacterial control, during pertussis infection. This implicates TREM-1 as a potential therapeutic target for the treatment of pertussis.
Collapse
|
22
|
Locht C. The Path to New Pediatric Vaccines against Pertussis. Vaccines (Basel) 2021; 9:vaccines9030228. [PMID: 33807962 PMCID: PMC7998139 DOI: 10.3390/vaccines9030228] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 12/21/2022] Open
Abstract
Whooping cough, or pertussis, mostly caused by Bordetella pertussis, is a respiratory disease that affects all age groups, but severe and fatal pertussis occurs almost exclusively in young children. The widespread use of whole-cell and, more recently, of acellular vaccines has substantially reduced the disease incidence. However, it has not been eliminated in any part of the world and has made a worrisome rebound in several areas. Cocoon and maternal immunization have been implemented in several countries but have their intrinsic limitations. To effectively control pertussis, novel vaccines are needed that protect against disease and prevent B. pertussis infection and transmission, which is not the case for current vaccines. Several approaches are contemplated, including alternative administration routes, such as nasal immunization, improvement of acellular vaccines by adding more antigens and T-cell-promoting adjuvants, and the development of novel vaccines, such as outer membrane vesicles and live attenuated vaccines. Among them, only a live attenuated vaccine has so far been assessed for safety and immunogenicity in preclinical models other than mice and is in clinical development. Before any of these vaccines can be used in neonates, extensive safety and immunogenicity assessment in pre-clinical neonatal models and in carefully designed clinical trials is necessary. The aim of this review is to discuss the current pertussis problem, implemented strategies to resolve it, the value of animal models and novel vaccine approaches.
Collapse
Affiliation(s)
- Camille Locht
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| |
Collapse
|
23
|
IL-17 mediates protective immunity against nasal infection with Bordetella pertussis by mobilizing neutrophils, especially Siglec-F + neutrophils. Mucosal Immunol 2021; 14:1183-1202. [PMID: 33976385 PMCID: PMC8379078 DOI: 10.1038/s41385-021-00407-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 03/26/2021] [Accepted: 04/15/2021] [Indexed: 02/04/2023]
Abstract
Understanding the mechanism of protective immunity in the nasal mucosae is central to the design of more effective vaccines that prevent nasal infection and transmission of Bordetella pertussis. We found significant infiltration of IL-17-secreting CD4+ tissue-resident memory T (TRM) cells and Siglec-F+ neutrophils into the nasal tissue during primary infection with B. pertussis. Il17A-/- mice had significantly higher bacterial load in the nasal mucosae, associated with significantly reduced infiltration of Siglec-F+ neutrophils. Re-infected convalescent mice rapidly cleared B. pertussis from the nasal cavity and this was associated with local expansion of IL-17-producing CD4+ TRM cells. Depletion of CD4 T cells from the nasal tissue during primary infection or after re-challenge of convalescent mice significantly delayed clearance of bacteria from the nasal mucosae. Protection was lost in Il17A-/- mice and this was associated with significantly less infiltration of Siglec-F+ neutrophils and antimicrobial peptide (AMP) production. Finally, depletion of neutrophils reduced the clearance of B. pertussis following re-challenge of convalescent mice. Our findings demonstrate that IL-17 plays a critical role in natural and acquired immunity to B. pertussis in the nasal mucosae and this effect is mediated by mobilizing neutrophils, especially Siglec-F+ neutrophils, which have high neutrophil extracellular trap (NET) activity.
Collapse
|
24
|
Blackwood CB, Sen-Kilic E, Boehm DT, Hall JM, Varney ME, Wong TY, Bradford SD, Bevere JR, Witt WT, Damron FH, Barbier M. Innate and Adaptive Immune Responses against Bordetella pertussis and Pseudomonas aeruginosa in a Murine Model of Mucosal Vaccination against Respiratory Infection. Vaccines (Basel) 2020; 8:vaccines8040647. [PMID: 33153066 PMCID: PMC7712645 DOI: 10.3390/vaccines8040647] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/23/2020] [Accepted: 10/28/2020] [Indexed: 12/26/2022] Open
Abstract
Whole cell vaccines are frequently the first generation of vaccines tested for pathogens and can inform the design of subsequent acellular or subunit vaccines. For respiratory pathogens, administration of vaccines at the mucosal surface can facilitate the generation of a localized mucosal immune response. Here, we examined the innate and vaccine-induced immune responses to infection by two respiratory pathogens: Bordetella pertussis and Pseudomonas aeruginosa. In a model of intranasal administration of whole cell vaccines (WCVs) with the adjuvant curdlan, we examined local and systemic immune responses following infection. These studies showed that intranasal vaccination with a WCV led to a reduction of the bacterial burden in the airways of animals infected with the respective pathogen. However, there were unique changes in the cytokines produced, cells recruited, and inflammation at the site of infection. Both mucosal vaccinations induced antibodies that bind the target pathogen, but linear regression and principal component analysis revealed that protection from these pathogens is not solely related to antibody titer. Protection from P. aeruginosa correlated to a reduction in lung weight, blood lymphocytes and neutrophils, and the cytokines IL-6, TNF-α, KC/GRO, and IL-10, and promotion of serum IgG antibodies and the cytokine IFN-γ in the lung. Protection from B. pertussis infection correlated strongly with increased anti-B-pertussis serum IgG antibodies. These findings reveal valuable correlates of protection for mucosal vaccination that can be used for further development of both B. pertussis and P. aeruginosa vaccines.
Collapse
|
25
|
Chasaide CN, Mills KH. Next-Generation Pertussis Vaccines Based on the Induction of Protective T Cells in the Respiratory Tract. Vaccines (Basel) 2020; 8:E621. [PMID: 33096737 PMCID: PMC7711671 DOI: 10.3390/vaccines8040621] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/13/2020] [Accepted: 10/16/2020] [Indexed: 12/11/2022] Open
Abstract
Immunization with current acellular pertussis (aP) vaccines protects against severe pertussis, but immunity wanes rapidly after vaccination and these vaccines do not prevent nasal colonization with Bordetella pertussis. Studies in mouse and baboon models have demonstrated that Th1 and Th17 responses are integral to protective immunity induced by previous infection with B. pertussis and immunization with whole cell pertussis (wP) vaccines. Mucosal Th17 cells, IL-17 and secretory IgA (sIgA) are particularly important in generating sustained sterilizing immunity in the nasal cavity. Current aP vaccines induce potent IgG and Th2-skewed T cell responses but are less effective at generating Th1 and Th17 responses and fail to prime respiratory tissue-resident memory T (TRM) cells, that maintain long-term immunity at mucosal sites. In contrast, a live attenuated pertussis vaccine, pertussis outer membrane vesicle (OMV) vaccines or aP vaccines formulated with novel adjuvants do induce cellular immune responses in the respiratory tract, especially when delivered by the intranasal route. An increased understanding of the mechanisms of sustained protective immunity, especially the role of respiratory TRM cells, will facilitate the development of next generation pertussis vaccines that not only protect against pertussis disease, but prevent nasal colonization and transmission of B. pertussis.
Collapse
Affiliation(s)
| | - Kingston H.G. Mills
- School of Biochemistry and Immunology, Trinity College Dublin, 2, D02 PN40 Dublin, Ireland;
| |
Collapse
|
26
|
Innate Lymphoid Cells: Important Regulators of Host-Bacteria Interaction for Border Defense. Microorganisms 2020; 8:microorganisms8091342. [PMID: 32887435 PMCID: PMC7563982 DOI: 10.3390/microorganisms8091342] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022] Open
Abstract
Innate lymphoid cells (ILCs) are a recently discovered type of innate immune lymphocyte. They include three different groups classified by the nature of the transcription factors required for their development and by the cytokines they produce. ILCs mainly reside in tissues close to the mucosal barrier such as the respiratory and gastrointestinal tracts. Due to their close proximity to the mucosal surface, ILCs are exposed to a variety of both commensal and pathogenic bacteria. Under non-pathological conditions, ILCs have been shown to be important regulators for the maintenance of tissue homeostasis by mutual interactions with the microbiome. Besides these important functions at homeostasis, several studies have also provided emerging evidence that ILCs contribute to defense against pathogenic bacterial infection by responding rapidly to the pathogens as well as orchestrating other immune cells. In this review, we summarize recent advances in our understanding of the interactions of ILCs and bacteria, with special focus on the function of the different ILC subsets in bacterial infections.
Collapse
|
27
|
Ardanuy J, Scanlon K, Skerry C, Fuchs SY, Carbonetti NH. Age-Dependent Effects of Type I and Type III IFNs in the Pathogenesis of Bordetella pertussis Infection and Disease. THE JOURNAL OF IMMUNOLOGY 2020; 204:2192-2202. [PMID: 32152071 PMCID: PMC7141952 DOI: 10.4049/jimmunol.1900912] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 02/14/2020] [Indexed: 12/20/2022]
Abstract
Type I and III IFNs play diverse roles in bacterial infections, being protective for some but deleterious for others. Using RNA-sequencing transcriptomics we investigated lung gene expression responses to Bordetella pertussis infection in adult mice, revealing that type I and III IFN pathways may play an important role in promoting inflammatory responses. In B. pertussis-infected mice, lung type I/III IFN responses correlated with increased proinflammatory cytokine expression and with lung inflammatory pathology. In mutant mice with increased type I IFN receptor (IFNAR) signaling, B. pertussis infection exacerbated lung inflammatory pathology, whereas knockout mice with defects in type I IFN signaling had lower levels of lung inflammation than wild-type mice. Curiously, B. pertussis-infected IFNAR1 knockout mice had wild-type levels of lung inflammatory pathology. However, in response to infection these mice had increased levels of type III IFN expression, neutralization of which reduced lung inflammation. In support of this finding, B. pertussis-infected mice with a knockout mutation in the type III IFN receptor (IFNLR1) and double IFNAR1/IFNLR1 knockout mutant mice had reduced lung inflammatory pathology compared with that in wild-type mice, indicating that type III IFN exacerbates lung inflammation. In marked contrast, infant mice did not upregulate type I or III IFNs in response to B. pertussis infection and were protected from lethal infection by increased type I IFN signaling. These results indicate age-dependent effects of type I/III IFN signaling during B. pertussis infection and suggest that these pathways represent targets for therapeutic intervention in pertussis.
Collapse
Affiliation(s)
- Jeremy Ardanuy
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201; and
| | - Karen Scanlon
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201; and
| | - Ciaran Skerry
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201; and
| | - Serge Y Fuchs
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104
| | - Nicholas H Carbonetti
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201; and
| |
Collapse
|
28
|
Commensal Microbes Affect Host Humoral Immunity to Bordetella pertussis Infection. Infect Immun 2019; 87:IAI.00421-19. [PMID: 31308086 PMCID: PMC6759300 DOI: 10.1128/iai.00421-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 06/11/2019] [Indexed: 01/07/2023] Open
Abstract
As important players in the host defense system, commensal microbes and the microbiota influence multiple aspects of host physiology. Bordetella pertussis infection is highly contagious among humans. However, the roles of the microbiota in B. pertussis pathogenesis are poorly understood. Here, we show that antibiotic-mediated depletion of the microbiota results in increased susceptibility to B. pertussis infection during the early stage. The increased susceptibility was associated with a marked impairment of the systemic IgG, IgG2a, and IgG1 antibody responses to B. pertussis infection after antibiotic treatment. Furthermore, the microbiota impacted the short-lived plasma cell responses as well as the recall responses of memory B cells to B. pertussis infection. Finally, we found that the dysbiosis caused by antibiotic treatment affects CD4+ T cell generation and PD-1 expression on CD4+ T cells and thereby perturbs plasma cell differentiation. Our results have revealed the importance of commensal microbes in modulating host immune responses to B. pertussis infection and support the possibility of controlling the severity of B. pertussis infection in humans by manipulating the microbiota.
Collapse
|
29
|
Kroes MM, Mariman R, Hijdra D, Hamstra HJ, van Boxtel KJWM, van Putten JPM, de Wit J, Pinelli E. Activation of Human NK Cells by Bordetella pertussis Requires Inflammasome Activation in Macrophages. Front Immunol 2019; 10:2030. [PMID: 31507615 PMCID: PMC6718514 DOI: 10.3389/fimmu.2019.02030] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 08/12/2019] [Indexed: 12/14/2022] Open
Abstract
Pertussis is a highly contagious respiratory infection caused by the bacterium Bordetella pertussis. Humans are the only known natural reservoir of B. pertussis. In mice, macrophages and NK cells have a key role in confining B. pertussis to the respiratory tract. However, the mechanisms underlying this process, particularly during human infections, remain unclear. Here we characterized the activation of human macrophages and NK cells in response to B. pertussis and unraveled the role of inflammasomes in this process. NLRP3 inflammasome activation by B. pertussis in human macrophage-like THP-1 cells and primary monocyte-derived macrophages (mo-MΦ) was shown by the visualization of ASC-speck formation, pyroptosis, and the secretion of caspase-mediated IL-1β and IL-18. In contrast to macrophages, stimulation of human CD56+CD3− NK cells by B. pertussis alone did not result in activation of these cells. However, co-culture of B. pertussis-stimulated mo-MΦ and autologous NK cells resulted in high amounts of IFNγ secretion and an increased frequency of IL-2Rα+ and HLA-DR+ NK cells, indicating NK cell activation. This activation was significantly reduced upon inhibition of inflammasome activity or blocking of IL-18 in the mo-MΦ/NK cell co-culture. Furthermore, we observed increased secretion of proinflammatory cytokines in the B. pertussis-stimulated mo-MΦ/NK co-culture compared to the mo-MΦ single culture. Our results demonstrate that B. pertussis induces inflammasome activation in human macrophages and that the IL-18 produced by these cells is required for the activation of human NK cells, which in turn enhances the pro-inflammatory response to this pathogen. Our data provides a better understanding of the underlying mechanisms involved in the induction of innate immune responses against B. pertussis. These findings contribute to the knowledge required for the development of improved intervention strategies to control this highly contagious disease.
Collapse
Affiliation(s)
- Michiel M Kroes
- Center for Immunology of Infectious Diseases and Vaccines, Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands.,Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Rob Mariman
- Center for Immunology of Infectious Diseases and Vaccines, Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Daniëlle Hijdra
- Center for Immunology of Infectious Diseases and Vaccines, Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Hendrik-Jan Hamstra
- Center for Immunology of Infectious Diseases and Vaccines, Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Karlijn J W M van Boxtel
- Center for Immunology of Infectious Diseases and Vaccines, Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Jos P M van Putten
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Jelle de Wit
- Center for Immunology of Infectious Diseases and Vaccines, Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Elena Pinelli
- Center for Immunology of Infectious Diseases and Vaccines, Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| |
Collapse
|
30
|
Jeljeli M, Guérin-El Khourouj V, Pédron B, Gressens P, Sibony O, Sterkers G. Ontogeny of cytokine responses to PHA from birth to adulthood. Pediatr Res 2019; 86:63-70. [PMID: 30928996 DOI: 10.1038/s41390-019-0383-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/01/2019] [Accepted: 03/04/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Altered production of cytokines is believed to contribute to early childhood susceptibility to infection. The aim of this study was to get further insight into the developmental patterns of cytokine responses from birth to adulthood. METHODS The expression levels of 13 cytokines were compared in the supernatants of phytohemaggluttinin (PHA)-stimulated whole blood from healthy neonates (cord blood, n = 8), infants ( < 1-year-old, n = 20), and school-aged children (3-15 y; n = 20). Five adults were used as reference. RESULTS While Th1, Th2, and Th17 cytokine levels increased progressively from birth to childhood (Mann-Whitney, p < 0.003), high IL-10 secretion at birth dropped to low adult levels in infants (p < 0.004) such that a negative correlation between IL-10 and Th1, Th2, and Th17 cytokine levels at birth (Spearman's correlation, r < -0.70, p < 0.01) converted to a positive correlation in infants (r > 0.60, p < 0.001). Finally, high IL-2, IL-7, and Granulocyte-Colony Stimulating factor (G-CSF) cytokine levels at birth decreased steadily over the first year of life (Mann-Whitney, p ≤ 0.001). CONCLUSION The most noticeable result of the study is the rapid shift from enhanced IL-10 secretion capacity at birth toward balanced IL-10/Th1/Th2/Th17 cytokine levels early in life. This change appears an essential precondition to fight pathogens and at the same time to avoid overwhelming inflammatory reactions.
Collapse
Affiliation(s)
- Mohamed Jeljeli
- Laboratory of Immunology, Robert-Debré Hospital, Assistance Publique-Hôpitaux de Paris AP-HP, 75019, Paris, France.,Univ. Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Valérie Guérin-El Khourouj
- Laboratory of Immunology, Robert-Debré Hospital, Assistance Publique-Hôpitaux de Paris AP-HP, 75019, Paris, France.,Univ. Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Béatrice Pédron
- Laboratory of Immunology, Robert-Debré Hospital, Assistance Publique-Hôpitaux de Paris AP-HP, 75019, Paris, France.,Univ. Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Pierre Gressens
- Univ. Paris Diderot, Sorbonne Paris Cité, Paris, France.,INSERM UMR1141, Paris, France
| | - Olivier Sibony
- Univ. Paris Diderot, Sorbonne Paris Cité, Paris, France.,Department of Gynecology Obstetric, Robert-Debré Hospital, Assistance Publique-Hôpitaux de Paris AP-HP, 75019, Paris, France
| | - Ghislaine Sterkers
- Laboratory of Immunology, Robert-Debré Hospital, Assistance Publique-Hôpitaux de Paris AP-HP, 75019, Paris, France. .,Univ. Paris Diderot, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
31
|
Wilk MM, Borkner L, Misiak A, Curham L, Allen AC, Mills KHG. Immunization with whole cell but not acellular pertussis vaccines primes CD4 T RM cells that sustain protective immunity against nasal colonization with Bordetella pertussis. Emerg Microbes Infect 2019; 8:169-185. [PMID: 30866771 PMCID: PMC6455184 DOI: 10.1080/22221751.2018.1564630] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Protective immunity wanes rapidly after immunization of children with acellular pertussis (aP) vaccines and these vaccines do not prevent nasal colonization or transmission of Bordetella pertussis in baboons. In this study, we examined the role of tissue-resident memory T (TRM) cells in persistent protective immunity induced by infection or immunization with aP and whole-cell pertussis (wP) vaccines in mice. Immunization of mice with a wP vaccine protected against lung and nasal colonization, whereas an aP vaccine failed to protect in the nose. IL-17 and IFN-γ-secreting CD69+CD4+ TRM cells were expanded in the lung and nasal tissue after B. pertussis challenge of mice immunized with wP, but not aP vaccines. However, previous infection induced the most persistent protection against nasal colonization and this correlated with potent induction of nasal tissue TRM cells, especially IL-17-secreting TRM cells. Blocking T cell migration to respiratory tissue during immunization with a wP vaccine impaired bacterial clearance, whereas transfer of TRM cells from convalescent or wP-immunized mice conferred protection to naïve mice. Our findings reveal that previous infection or wP vaccination are significantly more effective than aP vaccination in conferring persistent protective immunity against B. pertussis and that this is mediated by respiratory TRM cells.
Collapse
Affiliation(s)
- Mieszko M Wilk
- a Immune Regulation Research Group, School of Biochemistry and Immunology , Trinity Biomedical Sciences Institute, Trinity College Dublin , Dublin , Ireland
| | - Lisa Borkner
- a Immune Regulation Research Group, School of Biochemistry and Immunology , Trinity Biomedical Sciences Institute, Trinity College Dublin , Dublin , Ireland
| | - Alicja Misiak
- a Immune Regulation Research Group, School of Biochemistry and Immunology , Trinity Biomedical Sciences Institute, Trinity College Dublin , Dublin , Ireland
| | - Lucy Curham
- a Immune Regulation Research Group, School of Biochemistry and Immunology , Trinity Biomedical Sciences Institute, Trinity College Dublin , Dublin , Ireland
| | - Aideen C Allen
- a Immune Regulation Research Group, School of Biochemistry and Immunology , Trinity Biomedical Sciences Institute, Trinity College Dublin , Dublin , Ireland
| | - Kingston H G Mills
- a Immune Regulation Research Group, School of Biochemistry and Immunology , Trinity Biomedical Sciences Institute, Trinity College Dublin , Dublin , Ireland
| |
Collapse
|
32
|
Scanlon K, Skerry C, Carbonetti N. Association of Pertussis Toxin with Severe Pertussis Disease. Toxins (Basel) 2019; 11:toxins11070373. [PMID: 31252532 PMCID: PMC6669598 DOI: 10.3390/toxins11070373] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/17/2019] [Accepted: 06/20/2019] [Indexed: 12/26/2022] Open
Abstract
Pertussis, caused by respiratory tract infection with the bacterial pathogen Bordetella pertussis, has long been considered to be a toxin-mediated disease. Bacteria adhere and multiply extracellularly in the airways and release several toxins, which have a variety of effects on the host, both local and systemic. Predominant among these toxins is pertussis toxin (PT), a multi-subunit protein toxin that inhibits signaling through a subset of G protein-coupled receptors in mammalian cells. PT activity has been linked with severe and lethal pertussis disease in young infants and a detoxified version of PT is a common component of all licensed acellular pertussis vaccines. The role of PT in typical pertussis disease in other individuals is less clear, but significant evidence supporting its contribution to pathogenesis has been accumulated from animal model studies. In this review we discuss the evidence indicating a role for PT in pertussis disease, focusing on its contribution to severe pertussis in infants, modulation of immune and inflammatory responses to infection, and the characteristic paroxysmal cough of pertussis.
Collapse
Affiliation(s)
- Karen Scanlon
- Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Ciaran Skerry
- Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nicholas Carbonetti
- Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
33
|
Argondizo-Correia C, Rodrigues AKS, de Brito CA. Neonatal Immunity to Bordetella pertussis Infection and Current Prevention Strategies. J Immunol Res 2019; 2019:7134168. [PMID: 30882004 PMCID: PMC6387735 DOI: 10.1155/2019/7134168] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/22/2018] [Accepted: 01/03/2019] [Indexed: 01/10/2023] Open
Abstract
Bordetella pertussis is the bacterial agent of whooping cough, an infectious disease that is reemerging despite high vaccine coverage. Newborn children are the most affected, not only because they are too young to be vaccinated but also due to qualitative and quantitative differences in their immune system, which makes them more susceptible to infection and severe manifestations, leading to a higher mortality rate comparing to other groups. Until recently, prevention consisted of vaccinating children in the first year of life and the herd vaccination of people directly in touch with them, but the increase in cases demands more effective strategies that can overcome the developing immune response in early life and induce protection while children are most vulnerable.
Collapse
Affiliation(s)
- Carolina Argondizo-Correia
- Institute of Tropical Medicine, University of São Paulo, Dr. Enéas de Carvalho Aguiar Avenue 470 Jardim América, São Paulo, SP 05403-000, Brazil
- Immunology Centre, Adolfo Lutz Institute, Dr. Arnaldo Avenue 351 Cerqueira César, São Paulo, SP 01246-000, Brazil
| | - Ana Kelly Sousa Rodrigues
- Immunology Centre, Adolfo Lutz Institute, Dr. Arnaldo Avenue 351 Cerqueira César, São Paulo, SP 01246-000, Brazil
| | - Cyro Alves de Brito
- Institute of Tropical Medicine, University of São Paulo, Dr. Enéas de Carvalho Aguiar Avenue 470 Jardim América, São Paulo, SP 05403-000, Brazil
- Immunology Centre, Adolfo Lutz Institute, Dr. Arnaldo Avenue 351 Cerqueira César, São Paulo, SP 01246-000, Brazil
| |
Collapse
|
34
|
Abstract
Pertussis or whooping cough, mainly caused by Bordetella pertussis, is a severe respiratory disease that can affect all age groups but is most severe and can be life-threatening in young children. Vaccines against this disease are widely available since the 1950s. Despite high global vaccination coverage, the disease is not under control in any country, and its incidence is even increasing in several parts of the world. Epidemiological and experimental evidence has shown that the vaccines fail to prevent B. pertussis infection and transmission, although they are very effective in preventing disease. Given the high infection rate of B. pertussis, effective control of the disease likely requires prevention of infection and transmission in addition to protection against disease. With rare exceptions B. pertussis infections are restricted to the airways and do not usually disseminate beyond the respiratory epithelium. Therefore, protection at the level of the respiratory mucosa may be helpful for an improved control of pertussis. Yet, compared to systemic responses, mucosal immune responses have attracted relatively little attention in the context of pertussis vaccine development. In this review we summarize the available literature on the role of mucosal immunity in the prevention of B. pertussis infection. In contrast to vaccination, natural infection in humans and experimental infections in animals induce strong secretory IgA responses in the naso-pharynx and in the lungs. Several studies have shown that secretory IgA may be instrumental in the control of B. pertussis infection. Furthermore, studies in mouse models have revealed that B. pertussis infection, but not immunization with current acellular pertussis vaccines induces resident memory T cells, which may also contribute to protection against colonization by B. pertussis. As these resident memory T cells are long lived, vaccines that are able to induce them should provide long-lasting immunity. As of today, only one vaccine designed to induce potent mucosal immunity is in clinical development. This vaccine is a live attenuated B. pertussis strain delivered nasally in order to mimic the natural route of infection. Due to its ability to induce mucosal immunity it is expected that this approach will contribute to improved control of pertussis.
Collapse
Affiliation(s)
- Luis Solans
- Center of Infection and Immunity of Lille, Institut Pasteur de Lille, Lille, France
- Inserm U1019, Lille, France
- CNRS UMR8204, Lille, France
- Center for Infection and Immunity of Lille, Univ. Lille, Lille, France
| | - Camille Locht
- Center of Infection and Immunity of Lille, Institut Pasteur de Lille, Lille, France
- Inserm U1019, Lille, France
- CNRS UMR8204, Lille, France
- Center for Infection and Immunity of Lille, Univ. Lille, Lille, France
| |
Collapse
|
35
|
Lambert EE, Buisman AM, van Els CACM. Superior B. pertussis Specific CD4+ T-Cell Immunity Imprinted by Natural Infection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1183:81-98. [PMID: 31321753 DOI: 10.1007/5584_2019_405] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pertussis remains endemic in vaccinated populations due to waning of vaccine-induced immunity and insufficient interruption of transmission. Correlates of long-term protection against whooping cough remain elusive but increasing evidence from experimental models indicates that the priming of particular lineages of B. pertussis (Bp) specific CD4+ T cells is essential to control bacterial load. Critical hallmarks of these protective CD4+ T cell lineages in animals are suggested to be their differentiation profile as Th1 and Th17 cells and their tissue residency. These features seem optimally primed by previous infection but insufficiently or only partially by current vaccines. In this review, evidence is sought indicating whether infection also drives such superior Bp specific CD4+ T cell lineages in humans. We highlight key features of effector immunity downstream of Th1 and Th17 cell cytokines that explain clearing of primary Bp infections in naïve hosts, and effective prevention of infection in convalescent hosts during secondary challenge. Outstanding questions are put forward that need answers before correlates of human Bp infection-primed CD4+ T cell immunity can be used as benchmark for the development of improved pertussis vaccines.
Collapse
Affiliation(s)
- Eleonora E Lambert
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Anne-Marie Buisman
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Cécile A C M van Els
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands.
| |
Collapse
|
36
|
PERISCOPE: road towards effective control of pertussis. THE LANCET. INFECTIOUS DISEASES 2018; 19:e179-e186. [PMID: 30503084 DOI: 10.1016/s1473-3099(18)30646-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 10/02/2018] [Accepted: 10/12/2018] [Indexed: 12/28/2022]
Abstract
The resurgence and changing epidemiology of pertussis in high-income countries, the high infant mortality caused by pertussis in low-income countries, and the increasing morbidity in all age groups worldwide call for a concerted effort to both improve the current vaccines and develop new vaccines and vaccination strategies against pertussis. In this Personal View, we identify several key obstacles on the path to developing a durable solution for global control of pertussis. To systematically address these obstacles, the PERtussIS Correlates Of Protection Europe (PERISCOPE) Consortium was established in March, 2016. The objectives of this consortium are to increase scientific understanding of immunity to pertussis in humans induced by vaccines and infections, to identify biomarkers of protective immunity, and to generate technologies and infrastructure for the future development of improved pertussis vaccines. By working towards the accelerated licensure and implementation of novel, well tolerated, and effective pertussis vaccines, we hope to strengthen and stimulate further collaboration and transparency between the key stakeholders, including the public, the scientific community, public health institutes, regulatory authorities, and vaccine manufacturers.
Collapse
|
37
|
Sustained protective immunity against Bordetella pertussis nasal colonization by intranasal immunization with a vaccine-adjuvant combination that induces IL-17-secreting T RM cells. Mucosal Immunol 2018; 11:1763-1776. [PMID: 30127384 DOI: 10.1038/s41385-018-0080-x] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 07/17/2018] [Accepted: 08/03/2018] [Indexed: 02/04/2023]
Abstract
Current acellular pertussis (aP) vaccines induce strong antibody and Th2 responses but fail to protect against nasal colonization and transmission of Bordetella pertussis. Furthermore, immunity wanes rapidly after immunization. We have developed a novel adjuvant combination (called LP-GMP), comprising c-di-GMP, an intracellular receptor stimulator of interferon genes (STING) agonist, and LP1569, a TLR2 agonist from B. pertussis, which synergistically induces production of IFN-β, IL-12 and IL-23, and maturation of dendritic cells. Parenteral immunization of mice with an experimental aP vaccine formulated with LP-GMP promoted Th1 and Th17 responses and conferred protection against lung infection with B. pertussis. Intranasal immunization with the same aP vaccine-induced potent B. pertussis-specific Th17 responses and IL-17-secreting respiratory tissue-resident memory (TRM) CD4 T cells, and conferred a high level of protection against nasal colonization as well as lung infection, which was sustained for at least 10 months. Furthermore, long-term protection against nasal colonization with B. pertussis correlated with the number of IL-17-secreting TRM cells in nasal tissue. Our study has identified an approach for inducing IL-17-secreting TRM cells that sustain sterilizing immunity against nasal colonization of mice with B. pertussis, and could form the basis of a third generation pertussis vaccine for humans.
Collapse
|
38
|
Boehm DT, Hall JM, Wong TY, DiVenere AM, Sen-Kilic E, Bevere JR, Bradford SD, Blackwood CB, Elkins CM, DeRoos KA, Gray MC, Cooper CG, Varney ME, Maynard JA, Hewlett EL, Barbier M, Damron FH. Evaluation of Adenylate Cyclase Toxoid Antigen in Acellular Pertussis Vaccines by Using a Bordetella pertussis Challenge Model in Mice. Infect Immun 2018; 86:e00857-17. [PMID: 30012638 PMCID: PMC6204743 DOI: 10.1128/iai.00857-17] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 07/10/2018] [Indexed: 12/21/2022] Open
Abstract
Bordetella pertussis is the primary causative agent of pertussis (whooping cough), which is a respiratory infection that leads to a violent cough and can be fatal in infants. There is a need to develop more effective vaccines because of the resurgence of cases of pertussis in the United States since the switch from the whole-cell pertussis vaccines (wP) to the acellular pertussis vaccines (aP; diphtheria-tetanus-acellular-pertussis vaccine/tetanus-diphtheria-pertussis vaccine). Adenylate cyclase toxin (ACT) is a major virulence factor of B. pertussis that is (i) required for establishment of infection, (ii) an effective immunogen, and (iii) a protective antigen. The C-terminal repeats-in-toxin domain (RTX) of ACT is sufficient to induce production of toxin-neutralizing antibodies. In this study, we characterized the effectiveness of vaccines containing the RTX antigen against experimental murine infection with B. pertussis RTX was not protective as a single-antigen vaccine against B. pertussis challenge, and adding RTX to 1/5 human dose of aP did not enhance protection. Since the doses of aP used in murine studies are not proportionate to mouse/human body masses, we titrated the aP from 1/20 to 1/160 of the human dose. Mice receiving 1/80 human aP dose had bacterial burden comparable to those of naive controls. Adding RTX antigen to the 1/80 aP base resulted in enhanced bacterial clearance. Inclusion of RTX induced production of antibodies recognizing RTX, enhanced production of anti-pertussis toxin, decreased secretion of proinflammatory cytokines, such as interleukin-6, and decreased recruitment of total macrophages in the lung. This study shows that adding RTX antigen to an appropriate dose of aP can enhance protection against B. pertussis challenge in mice.
Collapse
Affiliation(s)
- Dylan T Boehm
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Jesse M Hall
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Ting Y Wong
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Andrea M DiVenere
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Emel Sen-Kilic
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Justin R Bevere
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Shelby D Bradford
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Catherine B Blackwood
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Cody M Elkins
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
| | - Katherine A DeRoos
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Mary C Gray
- Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - C Garret Cooper
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
- Department of Medicine, Section of Infectious Diseases, West Virginia University, Morgantown, West Virginia, USA
| | - Melinda E Varney
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - Jennifer A Maynard
- Department of Chemical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Erik L Hewlett
- Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - Mariette Barbier
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| | - F Heath Damron
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, West Virginia, USA
| |
Collapse
|
39
|
Differences in innate IFNγ and IL-17 responses to Bordetella pertussis between BALB/c and C57BL/6 mice: role of γδT cells, NK cells, and dendritic cells. Immunol Res 2018; 65:1139-1149. [PMID: 29052125 DOI: 10.1007/s12026-017-8957-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cell-mediated immune responses characterized by the secretion of IFNγ and IL-17 play an important role in the immune response to Bordetella pertussis (B. pertussis). We investigated innate sources of IFNγ and IL-17 upon stimulation of spleen cells from BALB/c (B/c) and C57BL/6 (B6) mice with heat-killed B. pertussis (hkBp). Spleen cells from B/c mice secreted less IFNγ and more IL-17 than those from B6 mice. Innate IFNγ was produced predominantly by NK cells in B/c mice and by CD8 T cells and NK cells in B6 mice. Innate IL-17 was produced primarily by γδT cells in both mouse strains. The secretion of IFNγ was abrogated by anti-IL-12, and the production of IL-17 was abolished by anti-IL-1β- and anti-IL23-neutralizing antibodies. B/c dendritic cells (DCs) stimulated with hkBp secreted significantly more IL-1β and less IL-12 than B6 DCs. Differences in JNK phosphorylation in DCs suggest that this pathway plays a role in the differences between B/c and B6 strains. Mixed cultures of DCs and γδT cells from B/c and B6 showed that cytokines from DCs as well as γδT cell-intrinsic factors contributed to the robust innate IL-17 response in B/c strain. Stimulation of γδT cells with IL-1β and IL-23 was sufficient for IL-17 secretion whereas IL-12 inhibited the secretion of IL-17. A larger fraction of γδT cells were γδT-17 cells in B/c mice than B6 mice. Our data indicate important roles for genetically determined factors in the innate IFNγ and IL-17 responses to B. pertussis.
Collapse
|
40
|
The Adjuvant Bordetella Colonization Factor A Attenuates Alum-Induced Th2 Responses and Enhances Bordetella pertussis Clearance from Mouse Lungs. Infect Immun 2018. [PMID: 29531137 DOI: 10.1128/iai.00935-17] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The reemergence of pertussis or whooping cough in several countries highlights the need for better vaccines. Acellular pertussis vaccines (aPV) contain alum as the adjuvant and elicit Th2-biased immune responses that are less effective in protecting against infection than the reactogenic whole-cell pertussis vaccines (wPV), which elicit primarily a Th1/Th17 response. An important goal for the field is to devise aPV that will induce immune responses similar to those of wPV. We show that Bordetella colonization factor A (BcfA), an outer membrane protein from Bordetella bronchiseptica, has strong adjuvant function and elicits cellular and humoral immune responses to heterologous and Bordetella pertussis antigens. Addition of BcfA to a commercial aPV resulted in greater reduction of B. pertussis numbers from the lungs than that elicited by aPV alone. The more-efficient pathogen clearance was accompanied by increased interleukin-17 (IL-17) and reduced IL-5 and an increased ratio of IgG2/IgG1 antibodies. Thus, our results suggest that BcfA improves aPV-induced responses by modifying the alum-induced Th2-biased aPV response toward Th1/Th17. A redesigned aPV containing BcfA may allow better control of pertussis reemergence by reshaping immune responses to resemble those elicited by wPV immunization.
Collapse
|
41
|
Torkashvand A, Bahrami F, Adib M, Ajdary S. Mucosal and systemic immune responses elicited by recombinant Lactococcus lactis expressing a fusion protein composed of pertussis toxin and filamentous hemagglutinin from Bordetella pertussis. Microb Pathog 2018; 120:155-160. [PMID: 29738814 PMCID: PMC7125623 DOI: 10.1016/j.micpath.2018.05.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 05/03/2018] [Accepted: 05/04/2018] [Indexed: 01/30/2023]
Abstract
We constructed a food-grade expression system harboring a F1S1 fusion protein of Bordetella pertussis to be produced in Lactococcus lactis NZ3900 as a new oral vaccine model against whooping cough, caused by B. pertussis. F1S1 was composed of N-terminally truncated S1 subunit of pertussis toxin and type I immunodominant domain of filamentous hemagglutinin which are both known as protective immunogens against pertussis. The recombinant L. lactis was administered via oral or intranasal routes to BALB/c mice and the related specific systemic and mucosal immune responses were then evaluated. The results indicated significantly higher levels of specific IgA in the lung extracts and IgG in sera of mucosally-immunized mice, compared to their controls. It was revealed that higher levels of IgG2a, compared to IgG1, were produced in all mucosally-immunized mice. Moreover, immunized mice developed Th1 responses with high levels of IFN-γ production by the spleen cells. These findings provide evidence for L. lactis to be used as a suitable vehicle for expression and delivery of F1S1 fusion protein to mucosa and induction of appropriate systemic and mucosal immune responses against pertussis. Lactococcus lactis was used for expression of fusion protein from Bordetella pertussis. BALB/c mice were immunized via oral or intranasal routes with recombinant L. lactis. Strong mucosal and Th1 systemic immune responses were developed. L. lactis is a suitable vehicle for expression and delivery of B. pertussis fusion protein.
Collapse
Affiliation(s)
- Ali Torkashvand
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Islamic Republic of Iran
| | - Fariborz Bahrami
- Department of Immunology, Pasteur Institute of Iran, 69 Pasteur Ave., Tehran, 13169-43551, Islamic Republic of Iran
| | - Minoo Adib
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Islamic Republic of Iran
| | - Soheila Ajdary
- Department of Immunology, Pasteur Institute of Iran, 69 Pasteur Ave., Tehran, 13169-43551, Islamic Republic of Iran.
| |
Collapse
|
42
|
Diavatopoulos DA, Edwards KM. What Is Wrong with Pertussis Vaccine Immunity? Why Immunological Memory to Pertussis Is Failing. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a029553. [PMID: 28289059 DOI: 10.1101/cshperspect.a029553] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Memory responses seen after whole-cell pertussis (wP) and acellular pertussis (aP) vaccine priming are different and reflect better long-term protection against pertussis disease seen with the whole-cell vaccines. Although acellular vaccines generate higher levels of antigen-specific IgG to the antigens included in the aP vaccines, there are many more pertussis antigens included in whole-cell vaccines. Acellular vaccine priming is associated with skewing of the immune response to a more Th2-like response, whereas whole-cell priming is associated with a Th1/Th17 response. Repeated booster doses of acellular vaccine in children primed with acellular vaccine has been shown to result in progressively shorter duration of protection against disease. This may be explained by the generation of higher levels of antigen-specific IgG4, which does not bind complement and leads to a suboptimal inflammatory response and impaired phagocytosis and antimicrobial defense. In contrast, whole-cell priming followed by aP vaccine boosters results in better opsonization, phagocytosis, and complement mediated killing through the preferential induction of IgG1.
Collapse
Affiliation(s)
- Dimitri A Diavatopoulos
- Laboratory of Pediatric Infectious Diseases, Radboud Center for Infectious Diseases, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Kathryn Margaret Edwards
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232.,Vanderbilt Vaccine Research Program, Vanderbilt University, Nashville, Tennessee 37332
| |
Collapse
|
43
|
CD71 + erythroid suppressor cells impair adaptive immunity against Bordetella pertussis. Sci Rep 2017; 7:7728. [PMID: 28798335 PMCID: PMC5552872 DOI: 10.1038/s41598-017-07938-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/05/2017] [Indexed: 01/03/2023] Open
Abstract
Infant’s immune system cannot control infection or respond to vaccination as efficiently as older individuals, a phenomenon that has been attributed to immunological immaturity. Recently, we challenged this notion and proposed the presence of actively immunosuppressive and physiologically enriched CD71+ erythroid cells in neonates. Here we utilized Bordetella pertussis, a common neonatal respiratory tract pathogen, as a proof of concept to investigate the role of these cells in adaptive immunity. We observed that CD71+ cells have distinctive immunosuppressive properties and prevent recruitment of immune cells to the mucosal site of infection. CD71+ cells ablation unleashed induction of B. pertussis-specific protective cytokines (IL-17 and IFN-γ) in the lungs and spleen upon re-infection or vaccination. We also found that CD71+ cells suppress systemic and mucosal B. pertussis-specific antibody responses. Enhanced antigen-specific adaptive immunity following CD71+ cells depletion increased resistance of mice to B. pertussis infection. Furthermore, we found that human cord blood CD71+ cells also suppress T and B cell functions in vitro. Collectively, these data provide important insight into the role of CD71+ erythroid cells in adaptive immunity. We anticipate our results will spark renewed investigation in modulating the function of these cells to enhance host defense to infections in newborns.
Collapse
|
44
|
Dunsmore G, Bozorgmehr N, Delyea C, Koleva P, Namdar A, Elahi S. Erythroid Suppressor Cells Compromise Neonatal Immune Response against Bordetella pertussis. THE JOURNAL OF IMMUNOLOGY 2017; 199:2081-2095. [PMID: 28779022 DOI: 10.4049/jimmunol.1700742] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/08/2017] [Indexed: 12/15/2022]
Abstract
Newborns are highly susceptible to infection. The underlying mechanism of neonatal infection susceptibility has generally been associated with neonatal immune cell immaturity. In this study, we challenged this notion and built upon our recent discovery that neonates are physiologically enriched with erythroid TER119+CD71+ cells (Elahi et al. 2013. Nature 504: 158-162). We have used Bordetella pertussis, a common neonatal respiratory tract infection, as a proof of concept to investigate the role of these cells in newborns. We found that CD71+ cells have distinctive immune-suppressive properties and suppress innate immune responses against B. pertussis infection. CD71+ cell ablation unleashed innate immune response and restored resistance to B. pertussis infection. In contrast, adoptive transfer of neonatal CD71+ cells into adult recipients impaired their innate immune response to B. pertussis infection. Enhanced innate immune response to B. pertussis was characterized by increased production of protective cytokines IFN-γ, TNF-α, and IL-12, as well as recruitment of NK cells, CD11b+, and CD11c+ cells in the lung. Neonatal and human cord blood CD71+ cells express arginase II, and this enzymatic activity inhibits phagocytosis of B. pertussis in vitro. Thus, our study challenges the notion that neonatal infection susceptibility is due to immune cell-intrinsic defects and instead highlights active immune suppression mediated by abundant CD71+ cells in the newborn. Our findings provide additional support for the novel theme in neonatal immunology that immunosuppression is essential to dampen robust immune responses in the neonate. We anticipate that our results will spark renewed investigation in modulating the function of these cells and developing novel strategies for enhancing host defense to infections in newborns.
Collapse
Affiliation(s)
- Garett Dunsmore
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada; and.,Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Najmeh Bozorgmehr
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada; and
| | - Cole Delyea
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada; and
| | - Petya Koleva
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada; and
| | - Afshin Namdar
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada; and
| | - Shokrollah Elahi
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada; and .,Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| |
Collapse
|
45
|
Wilk MM, Misiak A, McManus RM, Allen AC, Lynch MA, Mills KHG. Lung CD4 Tissue-Resident Memory T Cells Mediate Adaptive Immunity Induced by Previous Infection of Mice with Bordetella pertussis. THE JOURNAL OF IMMUNOLOGY 2017; 199:233-243. [PMID: 28533445 DOI: 10.4049/jimmunol.1602051] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 04/25/2017] [Indexed: 12/27/2022]
Abstract
Th1 and Th17 cells have an established role in protective immunity to Bordetella pertussis, but this evidence is based largely on peripheral T cells. There is emerging evidence that local tissue-resident memory T (TRM) cells that accumulate in tissue following mucosal infection may be crucial for long-term immunity. In this study, we examined the role of respiratory CD4 TRM cells in immunity to B. pertussis Natural immunity to B. pertussis induced by infection is considered long lasting and effective at preventing reinfection. Consistent with this, we found that convalescent mice rapidly cleared the bacteria after reinfection. Furthermore, CD4 T cells with a TRM cell phenotype (CD44+CD62L-CD69+ or CD44+CD62L-CD69+CD103+) accumulated in the lungs of mice during infection with B. pertussis and significantly expanded through local proliferation following reinfection. These CD4 TRM cells were B. pertussis specific and secreted IL-17 or IL-17 and IFN-γ. Treatment of mice with FTY720, which prevented migration of T and B cells from lymph nodes to the circulation, significantly exacerbated B. pertussis infection. This was associated with significantly reduced infiltration of central memory T cells and B cells into the lungs. However, the local expansion of TRM cells and the associated rapid clearance of the secondary infection were not affected by treatment with FTY720 before rechallenge. Moreover, adoptive transfer of lung CD4 TRM cells conferred protection in naive mice. Our findings reveal that Ag-specific CD4 TRM cells play a critical role in adaptive immunity against reinfection and memory induced by natural infection with B. pertussis.
Collapse
Affiliation(s)
- Mieszko M Wilk
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; and
| | - Alicja Misiak
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; and
| | - Róisín M McManus
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; and.,Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Aideen C Allen
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; and
| | - Marina A Lynch
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Kingston H G Mills
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; and
| |
Collapse
|
46
|
Protective Role of Passively Transferred Maternal Cytokines against Bordetella pertussis Infection in Newborn Piglets. Infect Immun 2017; 85:IAI.01063-16. [PMID: 28167667 DOI: 10.1128/iai.01063-16] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 01/27/2017] [Indexed: 11/20/2022] Open
Abstract
Maternal vaccination represents a potential strategy to protect both the mother and the offspring against life-threatening infections. This protective role has mainly been associated with antibodies, but the role of cell-mediated immunity, in particular passively transferred cytokines, is not well understood. Here, using a pertussis model, we have demonstrated that immunization of pregnant sows with heat-inactivated bacteria leads to induction of a wide range of cytokines (e.g., tumor necrosis factor alpha [TNF-α], gamma interferon [IFN-γ], interleukin-6 [IL-6], IL-8, and IL-12/IL-23p40) in addition to pertussis-specific antibodies. These cytokines can be detected in the sera and colostrum/milk of vaccinated sows and subsequently were detected at significant levels in the serum and bronchoalveolar lavage fluid of piglets born to vaccinated sows together with pertussis-specific antibodies. In contrast, active vaccination of newborn piglets with heat-inactivated bacteria induced high levels of specific IgG and IgA but no cytokines. Although the levels of antibodies in vaccinated piglets were comparable to those of passively transferred antibodies, no protection against Bordetella pertussis infection was observed. Thus, our results demonstrate that a combination of passively transferred cytokines and antibodies is crucial for disease protection. The presence of passively transferred cytokines/antibodies influences the cytokine secretion ability of splenocytes in the neonate, which provides novel evidence that maternal immunization can influence the newborn's cytokine milieu and may impact immune cell differentiation (e.g., Th1/Th2 phenotype). Therefore, these maternally derived cytokines may play an essential role both as mediators of early defense against infections and possibly as modulators of the immune repertoire of the offspring.
Collapse
|
47
|
Misiak A, Wilk MM, Raverdeau M, Mills KHG. IL-17-Producing Innate and Pathogen-Specific Tissue Resident Memory γδ T Cells Expand in the Lungs of Bordetella pertussis-Infected Mice. THE JOURNAL OF IMMUNOLOGY 2016; 198:363-374. [PMID: 27864475 DOI: 10.4049/jimmunol.1601024] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 10/20/2016] [Indexed: 12/14/2022]
Abstract
γδ T cells play a role in protective immunity to infection at mucosal surface, but also mediate pathology in certain autoimmune diseases through innate IL-17 production. Recent reports have suggested that γδ T cells can have memory analogous to conventional αβ T cells. In this study we have examined the role of γδ T cells in immunity to the respiratory pathogen Bordetella pertussis γδ T cells, predominantly Vγ4-γ1- cells, produced IL-17 in the lungs as early as 2 h after infection. The bacterial burden during primary infection was significantly enhanced and the induction of antimicrobial peptides was reduced in the absence of early IL-17. A second peak of γδ T cells is detected in the lungs 7-14 d after challenge and these γδ T cells were pathogen specific. γδ T cells, exclusively Vγ4, from the lungs of infected but not naive mice produced IL-17 in response to heat-killed B. pertussis in the presence of APC. Furthermore, γδ T cells from the lungs of mice reinfected with B. pertussis produced significantly more IL-17 than γδ T cells from infected unprimed mice. γδ T cells with a tissue resident memory T cell phenotype (CD69+CD103+) were expanded in the lungs during infection with B. pertussis and proliferated rapidly after rechallenge of convalescent mice. Our findings demonstrate that lung γδ T cells provide an early source of innate IL-17, which promotes antimicrobial peptide production, whereas pathogen-specific Vγ4 cells function in adaptive immunological memory against B. pertussis.
Collapse
Affiliation(s)
- Alicja Misiak
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Mieszko M Wilk
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Mathilde Raverdeau
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Kingston H G Mills
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
48
|
Li P, Asokanathan C, Liu F, Khaing KK, Kmiec D, Wei X, Song B, Xing D, Kong D. PLGA nano/micro particles encapsulated with pertussis toxoid (PTd) enhances Th1/Th17 immune response in a murine model. Int J Pharm 2016; 513:183-190. [PMID: 27586408 DOI: 10.1016/j.ijpharm.2016.08.059] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/18/2016] [Accepted: 08/28/2016] [Indexed: 10/21/2022]
Abstract
Poly(lactic-co-glycolic acid) (PLGA) based nano/micro particles were investigated as a potential vaccine platform for pertussis antigen. Presentation of pertussis toxoid as nano/micro particles (NP/MP) gave similar antigen-specific IgG responses in mice compared to soluble antigen. Notably, in cell line based assays, it was found that PLGA based nano/micro particles enhanced the phagocytosis of fluorescent antigen-nano/micro particles by J774.2 murine monocyte/macrophage cells compared to soluble antigen. More importantly, when mice were immunised with the antigen-nano/micro particles they significantly increased antigen-specific Th1 cytokines INF-γ and IL-17 secretion in splenocytes after in vitro re-stimulation with heat killed Bordetalla pertussis, indicating the induction of a Th1/Th17 response. Also, presentation of pertussis antigen in a NP/MP formulation is able to provide protection against respiratory infection in a murine model. Thus, the NP/MP formulation may provide an alternative to conventional acellular vaccines to achieve a more balanced Th1/Th2 immune response.
Collapse
Affiliation(s)
- Pan Li
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin Key Laboratory of Biomaterial Research, Tianjin 300192, China
| | - Catpagavalli Asokanathan
- Division of Bacteriology, National Institute for Biological Standards and Control (NIBSC), Blanche Lane, South Mimms, Hertfordshire, EN6 3QG, UK
| | - Fang Liu
- Department of Pharmacy, Pharmacology and Postgraduate Medicine, University of Hertfordshire, Hatfield, AL10 9AB, UK.
| | - Kyi Kyi Khaing
- Department of Pharmacy, Pharmacology and Postgraduate Medicine, University of Hertfordshire, Hatfield, AL10 9AB, UK
| | - Dorota Kmiec
- Division of Bacteriology, National Institute for Biological Standards and Control (NIBSC), Blanche Lane, South Mimms, Hertfordshire, EN6 3QG, UK
| | - Xiaoqing Wei
- Cardiff Institute of Tissue Engineering & Repair, School of Dentistry, Collegeof Biomedical and Life Sciences, Cardiff University, UK
| | - Bing Song
- Cardiff Institute of Tissue Engineering & Repair, School of Dentistry, Collegeof Biomedical and Life Sciences, Cardiff University, UK
| | - Dorothy Xing
- Division of Bacteriology, National Institute for Biological Standards and Control (NIBSC), Blanche Lane, South Mimms, Hertfordshire, EN6 3QG, UK
| | - Deling Kong
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin Key Laboratory of Biomaterial Research, Tianjin 300192, China.
| |
Collapse
|
49
|
Hoonakker ME, Verhagen LM, van der Maas L, Metz B, Uittenbogaard JP, van de Waterbeemd B, van Els CACM, van Eden W, Hendriksen CFM, Sloots A, Han WGH. Adaptive immune response to whole cell pertussis vaccine reflects vaccine quality: A possible complementation to the Pertussis Serological Potency test. Vaccine 2016; 34:4429-36. [PMID: 27452867 DOI: 10.1016/j.vaccine.2016.07.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 06/17/2016] [Accepted: 07/06/2016] [Indexed: 11/19/2022]
Abstract
Whole cell Bordetella pertussis (wP) vaccines are still used in many countries to protect against the respiratory disease pertussis. The potency of whole-cell pertussis vaccine lots is determined by an intracerebral challenge test (the Kendrick test). This test is criticized due to lack of immunological relevance of the read-out after an intracerebral challenge with B. pertussis. The alternative in vivo test, which assesses specific antibody levels in serum after wP vaccination, is the Pertussis Serological Potency test (PSPT). Although the PSPT focuses on a parameter that contributes to protection, the protective immune mechanisms after wP vaccination includes more elements than specific antibody responses only. In this study, additional parameters were investigated, i.e. circulating pro-inflammatory cytokines, antibody specificity and T helper cell responses and it was evaluated whether they can be used as complementary readout parameters in the PSPT to assess wP lot quality. By deliberate manipulation of the vaccine preparation procedure, a panel of high, intermediate and low quality wP vaccines were made. The results revealed that these vaccines induced similar IL-6 and IP10 levels in serum 4h after vaccination (innate responses) and similar antibody levels directed against the entire bacterium. In contrast, the induced antibody specificity to distinct wP antigens differed after vaccination with high, intermediate and low quality wP vaccines. In addition, the magnitude of wP-induced Th cell responses (Th17, Th1 and Th2) was reduced after vaccination with a wP vaccine of low quality. T cell responses and antibody specificity are therefore correlates of qualitative differences in the investigated vaccines, while the current parameter of the PSPT alone was not sensitive enough to distinguish between vaccines of different qualities. This study demonstrates that assessment of the magnitude of Th cell responses and the antigen specificity of antibodies induced by wP vaccination could form valuable complementary parameters to the PSPT.
Collapse
Affiliation(s)
- M E Hoonakker
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands; Utrecht University, Faculty of Veterinary Medicine, Department Animals in Science and Society, Utrecht, The Netherlands.
| | - L M Verhagen
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands; Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - L van der Maas
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
| | - B Metz
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
| | - J P Uittenbogaard
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
| | - B van de Waterbeemd
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
| | - C A C M van Els
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - W van Eden
- Utrecht University, Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht, The Netherlands
| | - C F M Hendriksen
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands; Utrecht University, Faculty of Veterinary Medicine, Department Animals in Science and Society, Utrecht, The Netherlands
| | - A Sloots
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
| | - W G H Han
- Centre for Immunology of Infectious Diseases and Vaccines, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| |
Collapse
|
50
|
Pertussis specific cell-mediated immune responses ten years after acellular pertussis booster vaccination in young adults. Vaccine 2016; 34:341-9. [DOI: 10.1016/j.vaccine.2015.11.049] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 10/22/2015] [Accepted: 11/17/2015] [Indexed: 11/24/2022]
|