1
|
Saeland E, van der Fits L, Bolder R, Heemskerk-van der Meer M, Drijver J, van Polanen Y, Vaneman C, Tettero L, Cox F, Serroyen J, Jorgensen MJ, Langedijk JPM, Schuitemaker H, Callendret B, Zahn RC. Combination Ad26.RSV.preF/preF protein vaccine induces superior protective immunity compared with individual vaccine components in preclinical models. NPJ Vaccines 2023; 8:45. [PMID: 36949051 PMCID: PMC10033289 DOI: 10.1038/s41541-023-00637-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 02/27/2023] [Indexed: 03/24/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of severe respiratory disease for which no licensed vaccine is available. We have previously shown that a prefusion (preF) conformation-stabilized RSV F protein antigen and an adenoviral vector encoding RSV preF protein (Ad26.RSV.preF) are immunogenic and protective in animals when administered as single components. Here, we evaluated a combination of the 2 components, administered as a single injection. Strong induction of both humoral and cellular responses was shown in RSV-naïve and pre-exposed mice and pre-exposed African green monkeys (AGMs). Both components of the combination vaccine contributed to humoral immune responses, while the Ad26.RSV.preF component was the main contributor to cellular immune responses in both mice and AGMs. Immunization with the combination elicited superior protection against RSV A2 challenge in cotton rats. These results demonstrate the advantage of a combination vaccine and support further clinical development.
Collapse
Affiliation(s)
- Eirikur Saeland
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands.
| | | | - Renske Bolder
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| | | | - Joke Drijver
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| | | | | | | | - Freek Cox
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| | - Jan Serroyen
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| | - Matthew J Jorgensen
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | | | | | - Roland C Zahn
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| |
Collapse
|
2
|
Kumar NP, Padmapriyadarsini C, Rajamanickam A, Bhavani PK, Nancy A, Jayadeepa B, Selvaraj N, Asokan D, Renji RM, Venkataramani V, Tripathy S, Babu S. BCG vaccination induces enhanced frequencies of memory T cells and altered plasma levels of common γc cytokines in elderly individuals. PLoS One 2021; 16:e0258743. [PMID: 34758029 PMCID: PMC8580239 DOI: 10.1371/journal.pone.0258743] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 09/26/2021] [Indexed: 12/03/2022] Open
Abstract
BCG vaccination is known to induce innate immune memory, which confers protection against heterologous infections. However, the effect of BCG vaccination on the conventional adaptive immune cells subsets is not well characterized. We investigated the impact of BCG vaccination on the frequencies of T cell subsets and common gamma c (γc) cytokines in a group of healthy elderly individuals (age 60–80 years) at one month post vaccination as part of our clinical study to examine the effect of BCG on COVID-19. Our results demonstrate that BCG vaccination induced enhanced frequencies of central (p<0.0001) and effector memory (p<0.0001) CD4+ T cells and diminished frequencies of naïve (p<0.0001), transitional memory (p<0.0001), stem cell memory (p = 0.0001) CD4+ T cells and regulatory T cells. In addition, BCG vaccination induced enhanced frequencies of central (p = 0.0008), effector (p<0.0001) and terminal effector memory (p<0.0001) CD8+ T cells and diminished frequencies of naïve (p<0.0001), transitional memory (p<0.0001) and stem cell memory (p = 0.0034) CD8+T cells. BCG vaccination also induced enhanced plasma levels of IL-7 (p<0.0001) and IL-15 (p = 0.0020) but diminished levels of IL-2 (p = 0.0033) and IL-21 (p = 0.0020). Thus, BCG vaccination was associated with enhanced memory T cell subsets as well as memory enhancing γc cytokines in elderly individuals, suggesting its ability to induce non-specific adaptive immune responses.
Collapse
Affiliation(s)
- Nathella Pavan Kumar
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
- * E-mail: (NPK); (SB)
| | | | | | - Perumal Kannabiran Bhavani
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
| | - Arul Nancy
- ICMR-National Institute for Research in Tuberculosis, Chennai, India
| | - Bharathi Jayadeepa
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
| | - Nandhini Selvaraj
- ICMR-National Institute for Research in Tuberculosis, Chennai, India
| | - Dinesh Asokan
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
| | | | | | - Srikanth Tripathy
- ICMR-National Institute for Research in Tuberculosis-International Center for Excellence in Research, Chennai, India
- Dr D Y Patil Medical College, Hospital and Research Centre, Pune, India
| | - Subash Babu
- ICMR-National Institute for Research in Tuberculosis, Chennai, India
- * E-mail: (NPK); (SB)
| |
Collapse
|
3
|
Tomas J, Koo Y, Popoff D, Arce-Gorvel V, Hanniffy S, Gorvel JP, Mionnet C. PTX Instructs the Development of Lung-Resident Memory T Cells in Bordetella pertussis Infected Mice. Toxins (Basel) 2021; 13:toxins13090632. [PMID: 34564636 PMCID: PMC8470914 DOI: 10.3390/toxins13090632] [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: 06/16/2021] [Revised: 08/13/2021] [Accepted: 08/30/2021] [Indexed: 11/16/2022] Open
Abstract
Whooping cough is a severe, highly contagious disease of the human respiratory tract, caused by Bordetellapertussis. The pathogenicity requires several virulence factors, including pertussis toxin (PTX), a key component of current available vaccines. Current vaccines do not induce mucosal immunity. Tissue-resident memory T cells (Trm) are among the first lines of defense against invading pathogens and are involved in long-term protection. However, the factors involved in Trm establishment remain unknown. Comparing two B.pertussis strains expressing PTX (WT) or not (ΔPTX), we show that the toxin is required to generate both lung CD4+ and CD8+ Trm. Co-administering purified PTX with ΔPTX is sufficient to generate these Trm subsets. Importantly, adoptive transfer of lung CD4+ or CD8+ Trm conferred protection against B. pertussis in naïve mice. Taken together, our data demonstrate for the first time a critical role for PTX in the induction of mucosal long-term protection against B. pertussis.
Collapse
Affiliation(s)
- Julie Tomas
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Centre National de la Recherche Scientifique (CNRS), UMR7280, Parc Scientifique et Technologique de Luminy, Case 906, 13288 Marseille, France; (J.T.); (Y.K.); (D.P.); (V.A.-G.); (S.H.); (J.-P.G.)
| | - Yoon Koo
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Centre National de la Recherche Scientifique (CNRS), UMR7280, Parc Scientifique et Technologique de Luminy, Case 906, 13288 Marseille, France; (J.T.); (Y.K.); (D.P.); (V.A.-G.); (S.H.); (J.-P.G.)
- Laboratoire Adhesion & Inflammation, UMR INSERM 1067, UMR CNRS 7333, Aix-Marseille Université Case 937, CEDEX 09, 13288 Marseille, France
| | - Dimitri Popoff
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Centre National de la Recherche Scientifique (CNRS), UMR7280, Parc Scientifique et Technologique de Luminy, Case 906, 13288 Marseille, France; (J.T.); (Y.K.); (D.P.); (V.A.-G.); (S.H.); (J.-P.G.)
| | - Vilma Arce-Gorvel
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Centre National de la Recherche Scientifique (CNRS), UMR7280, Parc Scientifique et Technologique de Luminy, Case 906, 13288 Marseille, France; (J.T.); (Y.K.); (D.P.); (V.A.-G.); (S.H.); (J.-P.G.)
| | - Sean Hanniffy
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Centre National de la Recherche Scientifique (CNRS), UMR7280, Parc Scientifique et Technologique de Luminy, Case 906, 13288 Marseille, France; (J.T.); (Y.K.); (D.P.); (V.A.-G.); (S.H.); (J.-P.G.)
| | - Jean-Pierre Gorvel
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Centre National de la Recherche Scientifique (CNRS), UMR7280, Parc Scientifique et Technologique de Luminy, Case 906, 13288 Marseille, France; (J.T.); (Y.K.); (D.P.); (V.A.-G.); (S.H.); (J.-P.G.)
| | - Cyrille Mionnet
- Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University, UM2, Institut National de la Santé et de la Recherche Médicale (INSERM), U1104, Centre National de la Recherche Scientifique (CNRS), UMR7280, Parc Scientifique et Technologique de Luminy, Case 906, 13288 Marseille, France; (J.T.); (Y.K.); (D.P.); (V.A.-G.); (S.H.); (J.-P.G.)
- Correspondence:
| |
Collapse
|
4
|
George JA, AlShamsi SH, Alhammadi MH, Alsuwaidi AR. Exacerbation of Influenza A Virus Disease Severity by Respiratory Syncytial Virus Co-Infection in a Mouse Model. Viruses 2021; 13:v13081630. [PMID: 34452495 PMCID: PMC8402720 DOI: 10.3390/v13081630] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/16/2021] [Accepted: 08/16/2021] [Indexed: 01/15/2023] Open
Abstract
Influenza A virus (IAV) and respiratory syncytial virus (RSV) are leading causes of childhood infections. RSV and influenza are competitive in vitro. In this study, the in vivo effects of RSV and IAV co-infection were investigated. Mice were intranasally inoculated with RSV, with IAV, or with both viruses (RSV+IAV and IAV+RSV) administered sequentially, 24 h apart. On days 3 and 7 post-infection, lung tissues were processed for viral loads and immune cell populations. Lung functions were also evaluated. Mortality was observed only in the IAV+RSV group (50% of mice did not survive beyond 7 days). On day 3, the viral loads in single-infected and co-infected mice were not significantly different. However, on day 7, the IAV titer was much higher in the IAV+RSV group, and the RSV viral load was reduced. CD4 T cells were reduced in all groups on day 7 except in single-infected mice. CD8 T cells were higher in all experimental groups except the RSV-alone group. Increased airway resistance and reduced thoracic compliance were demonstrated in both co-infected groups. This model indicates that, among all the infection types we studied, infection with IAV followed by RSV is associated with the highest IAV viral loads and the most morbidity and mortality.
Collapse
Affiliation(s)
- Junu A. George
- Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates;
| | - Shaikha H. AlShamsi
- Department of Medical Education, Sheikh Khalifa Medical City, Abu Dhabi Health Services Company (SEHA), Abu Dhabi 51900, United Arab Emirates;
| | - Maryam H. Alhammadi
- Department of Medical Affairs, Sheikh Shakhbout Medical City, Abu Dhabi Health Services Company (SEHA), Abu Dhabi 11001, United Arab Emirates;
| | - Ahmed R. Alsuwaidi
- Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates;
- Correspondence:
| |
Collapse
|
5
|
Batista da Silva Galdino A, do Nascimento Rangel AH, Buttar HS, Sales Lima Nascimento M, Cristina Gavioli E, Oliveira RDP, Cavalcanti Sales D, Urbano SA, Anaya K. Bovine colostrum: benefits for the human respiratory system and potential contributions for clinical management of COVID-19. FOOD AGR IMMUNOL 2021. [DOI: 10.1080/09540105.2021.1892594] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Alyne Batista da Silva Galdino
- Unidade Acadêmica Especializada em Ciências Agrárias – UAECA, Universidade Federal do Rio Grande do Norte – UFRN, Macaíba, Brasil
| | | | - Harpal Singh Buttar
- Department of Pathology and Laboratory Medicine, University of Ottawa, School of Medicine, Ottawa, Canada
| | - Manuela Sales Lima Nascimento
- Departamento de Microbiologia e Parasitologia, Centro de Biociências, Universidade Federal do Rio Grande do Norte – UFRN, Natal, Brasil
| | - Elaine Cristina Gavioli
- Departamento de Biofísica e Farmacologia, Centro de Biociências, Universidade Federal do Rio Grande do Norte – UFRN, Natal, Brasil
| | - Riva de Paula Oliveira
- Departamento de Ciências Biológicas, Centro de Biociências, Universidade Federal do Rio Grande do Norte – UFRN, Natal, Brasil
| | - Danielle Cavalcanti Sales
- Unidade Acadêmica Especializada em Ciências Agrárias – UAECA, Universidade Federal do Rio Grande do Norte – UFRN, Macaíba, Brasil
| | - Stela Antas Urbano
- Unidade Acadêmica Especializada em Ciências Agrárias – UAECA, Universidade Federal do Rio Grande do Norte – UFRN, Macaíba, Brasil
| | - Katya Anaya
- Faculdade de Ciências da Saúde do Trairi – FACISA, Universidade Federal do Rio Grande do Norte – UFRN, Santa Cruz, Brasil
| |
Collapse
|
6
|
Immunogenicity and inflammatory properties of respiratory syncytial virus attachment G protein in cotton rats. PLoS One 2021; 16:e0246770. [PMID: 33600439 PMCID: PMC7891763 DOI: 10.1371/journal.pone.0246770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 01/25/2021] [Indexed: 12/25/2022] Open
Abstract
Human respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infection in infants and young children worldwide. The attachment (G) protein of RSV is synthesized by infected cells in both a membrane bound (mG) and secreted form (sG) and uses a CX3C motif for binding to its cellular receptor. Cell culture and mouse studies suggest that the G protein mimics the cytokine CX3CL1 by binding to CX3CR1 on immune cells, which is thought to cause increased pulmonary inflammation in vivo. However, because these studies have used RSV lacking its G protein gene or blockade of the G protein with a G protein specific monoclonal antibody, the observed reduction in inflammation may be due to reduced virus replication and spread, and not to a direct role for G protein as a viral chemokine. In order to more directly determine the influence of the soluble and the membrane-bound forms of G protein on the immune system independent of its attachment function for the virion, we expressed the G protein in cotton rat lungs using adeno-associated virus (AAV), a vector system which does not itself induce inflammation. We found no increase in pulmonary inflammation as determined by histology and bronchoalveolar lavage after inoculation of AAVs expressing the membrane bound G protein, the secreted G protein or the complete G protein gene which expresses both forms. The long-term low-level expression of AAV-G did, however, result in the induction of non-neutralizing antibodies, CD8 T cells and partial protection from challenge with RSV. Complete protection was accomplished through co-immunization with AAV-G and an AAV expressing cotton rat interferon α.
Collapse
|
7
|
Gustafson CE, Kim C, Weyand CM, Goronzy JJ. Influence of immune aging on vaccine responses. J Allergy Clin Immunol 2020; 145:1309-1321. [PMID: 32386655 PMCID: PMC7198995 DOI: 10.1016/j.jaci.2020.03.017] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/20/2020] [Accepted: 03/24/2020] [Indexed: 12/14/2022]
Abstract
Impaired vaccine responses in older individuals are associated with alterations in both the quantity and quality of the T-cell compartment with age. As reviewed herein, the T-cell response to vaccination requires a fine balance between the generation of inflammatory effector T cells versus follicular helper T (TFH) cells that mediate high-affinity antibody production in tandem with the induction of long-lived memory cells for effective recall immunity. During aging, we find that this balance is tipped where T cells favor short-lived effector but not memory or TFH responses. Consistently, vaccine-induced antibodies commonly display a lower protective capacity. Mechanistically, multiple, potentially targetable, changes in T cells have been identified that contribute to these age-related defects, including posttranscription regulation, T-cell receptor signaling, and metabolic function. Although research into the induction of tissue-specific immunity by vaccines and with age is still limited, current mechanistic insights provide a framework for improved design of age-specific vaccination strategies that require further evaluation in a clinical setting.
Collapse
Affiliation(s)
- Claire E Gustafson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, Calif; Department of Medicine, Veterans Administration Healthcare System, Palo Alto, Calif
| | - Chulwoo Kim
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, Calif; Department of Medicine, Veterans Administration Healthcare System, Palo Alto, Calif
| | - Cornelia M Weyand
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, Calif; Department of Medicine, Veterans Administration Healthcare System, Palo Alto, Calif
| | - Jörg J Goronzy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, Calif; Department of Medicine, Veterans Administration Healthcare System, Palo Alto, Calif.
| |
Collapse
|
8
|
Pfeffer PE, Ho TR, Mann EH, Kelly FJ, Sehlstedt M, Pourazar J, Dove RE, Sandstrom T, Mudway IS, Hawrylowicz CM. Urban particulate matter stimulation of human dendritic cells enhances priming of naive CD8 T lymphocytes. Immunology 2018; 153:502-512. [PMID: 29044495 PMCID: PMC5838419 DOI: 10.1111/imm.12852] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 10/10/2017] [Accepted: 10/11/2017] [Indexed: 02/06/2023] Open
Abstract
Epidemiological studies have consistently shown associations between elevated concentrations of urban particulate matter (UPM) air pollution and exacerbations of asthma and chronic obstructive pulmonary disease, which are both associated with viral respiratory infections. The effects of UPM on dendritic cell (DC) -stimulated CD4 T lymphocytes have been investigated previously, but little work has focused on CD8 T-lymphocyte responses despite their importance in anti-viral immunity. To address this, we examined the effects of UPM on DC-stimulated naive CD8 T-cell responses. Expression of the maturation/activation markers CD83, CCR7, CD40 and MHC class I on human myeloid DCs (mDCs) was characterized by flow cytometry after stimulation with UPMin vitro in the presence/absence of granulocyte-macrophage colony-stimulating factor (GM-CSF). The capacity of these mDCs to stimulate naive CD8 T-lymphocyte responses in allogeneic co-culture was then assessed by measuring T-cell cytokine secretion using cytometric bead array, and proliferation and frequency of interferon-γ (IFN-γ)-producing T lymphocytes by flow cytometry. Treatment of mDCs with UPM increased expression of CD83 and CCR7, but not MHC class I. In allogeneic co-cultures, UPM treatment of mDCs enhanced CD8 T-cell proliferation and the frequency of IFN-γ+ cells. The secretion of tumour necrosis factor-α, interleukin-13, Granzyme A and Granzyme B were also increased. GM-CSF alone, and in concert with UPM, enhanced many of these T-cell functions. The PM-induced increase in Granzyme A was confirmed in a human experimental diesel exposure study. These data demonstrate that UPM treatment of mDCs enhances priming of naive CD8 T lymphocytes and increases production of pro-inflammatory cytokines. Such UPM-induced stimulation of CD8 cells may potentiate T-lymphocyte cytotoxic responses upon concurrent airway infection, increasing bystander damage to the airways.
Collapse
Affiliation(s)
- Paul E. Pfeffer
- MRC and Asthma UK Centre for Allergic Mechanisms of AsthmaKing's College LondonGuy's HospitalLondonUK
- Present address:
William Harvey Research InstituteQueen Mary University of LondonLondonEC1M 6BQUK
| | - Tzer R. Ho
- MRC and Asthma UK Centre for Allergic Mechanisms of AsthmaKing's College LondonGuy's HospitalLondonUK
| | - Elizabeth H. Mann
- MRC and Asthma UK Centre for Allergic Mechanisms of AsthmaKing's College LondonGuy's HospitalLondonUK
| | - Frank J. Kelly
- MRC and Asthma UK Centre for Allergic Mechanisms of AsthmaKing's College LondonGuy's HospitalLondonUK
- Environmental Research GroupMRC‐PHE Centre for Environment and HealthKing's College LondonLondonUK
- NIHR Health Protection Research Unit in Health Impact of Environmental HazardsFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Maria Sehlstedt
- Division of MedicineDepartment of Public Health and Clinical MedicineUmeå UniversityUmeåSweden
| | - Jamshid Pourazar
- Division of MedicineDepartment of Public Health and Clinical MedicineUmeå UniversityUmeåSweden
| | - Rosamund E. Dove
- Environmental Research GroupMRC‐PHE Centre for Environment and HealthKing's College LondonLondonUK
- NIHR Health Protection Research Unit in Health Impact of Environmental HazardsFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Thomas Sandstrom
- Division of MedicineDepartment of Public Health and Clinical MedicineUmeå UniversityUmeåSweden
| | - Ian S. Mudway
- Environmental Research GroupMRC‐PHE Centre for Environment and HealthKing's College LondonLondonUK
- NIHR Health Protection Research Unit in Health Impact of Environmental HazardsFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Catherine M. Hawrylowicz
- MRC and Asthma UK Centre for Allergic Mechanisms of AsthmaKing's College LondonGuy's HospitalLondonUK
- NIHR Health Protection Research Unit in Health Impact of Environmental HazardsFaculty of Life Sciences and MedicineKing's College LondonLondonUK
| |
Collapse
|
9
|
González AE, Lay MK, Jara EL, Espinoza JA, Gómez RS, Soto J, Rivera CA, Abarca K, Bueno SM, Riedel CA, Kalergis AM. Aberrant T cell immunity triggered by human Respiratory Syncytial Virus and human Metapneumovirus infection. Virulence 2016; 8:685-704. [PMID: 27911218 DOI: 10.1080/21505594.2016.1265725] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Human Respiratory syncytial virus (hRSV) and human metapneumovirus (hMPV) are the two major etiological viral agents of lower respiratory tract diseases, affecting mainly infants, young children and the elderly. Although the infection of both viruses trigger an antiviral immune response that mediate viral clearance and disease resolution in immunocompetent individuals, the promotion of long-term immunity appears to be deficient and reinfection are common throughout life. A possible explanation for this phenomenon is that hRSV and hMPV, can induce aberrant T cell responses, which leads to exacerbated lung inflammation and poor T and B cell memory immunity. The modulation of immune response exerted by both viruses include different strategies such as, impairment of immunological synapse mediated by viral proteins or soluble factors, and the induction of pro-inflammatory cytokines by epithelial cells, among others. All these viral strategies contribute to the alteration of the adaptive immunity in order to increase the susceptibility to reinfections. In this review, we discuss current research related to the mechanisms underlying the impairment of T and B cell immune responses induced by hRSV and hMPV infection. In addition, we described the role each virulence factor involved in immune modulation caused by these viruses.
Collapse
Affiliation(s)
- Andrea E González
- a Millennium Institute of Immunology and Immunotherapy , Departamento de Genética Molecular y Microbiología , Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Margarita K Lay
- b Departamento de Biotecnología , Facultad de Ciencias del Mar y Recursos Biológicos, Universidad de Antofagasta , Antofagasta , Chile
| | - Evelyn L Jara
- a Millennium Institute of Immunology and Immunotherapy , Departamento de Genética Molecular y Microbiología , Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Janyra A Espinoza
- a Millennium Institute of Immunology and Immunotherapy , Departamento de Genética Molecular y Microbiología , Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Roberto S Gómez
- a Millennium Institute of Immunology and Immunotherapy , Departamento de Genética Molecular y Microbiología , Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Jorge Soto
- a Millennium Institute of Immunology and Immunotherapy , Departamento de Genética Molecular y Microbiología , Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Claudia A Rivera
- a Millennium Institute of Immunology and Immunotherapy , Departamento de Genética Molecular y Microbiología , Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Katia Abarca
- c Departamento de Pediatría , Facultad de Medicina, Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Susan M Bueno
- a Millennium Institute of Immunology and Immunotherapy , Departamento de Genética Molecular y Microbiología , Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile , Santiago , Chile.,d INSERM UMR1064 , Nantes , France
| | - Claudia A Riedel
- e Millennium Institute of Immunology and Immunotherapy , Departamento de Ciencias Biológicas , Facultad de Ciencias Biológicas y Facultad de Medicina, Universidad Andrés Bello , Santiago , Chile
| | - Alexis M Kalergis
- a Millennium Institute of Immunology and Immunotherapy , Departamento de Genética Molecular y Microbiología , Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile , Santiago , Chile.,c Departamento de Pediatría , Facultad de Medicina, Pontificia Universidad Católica de Chile , Santiago , Chile.,f Millennium Institute of Immunology and Immunotherapy , Departamento de Endocrinología , Facultad de Medicina, Pontificia Universidad Católica de Chile , Santiago , Chile
| |
Collapse
|
10
|
Superior isolation of antigen-specific brain infiltrating T cells using manual homogenization technique. J Immunol Methods 2016; 439:23-28. [PMID: 27623324 DOI: 10.1016/j.jim.2016.09.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 09/02/2016] [Accepted: 09/06/2016] [Indexed: 11/23/2022]
Abstract
Effective recovery of activated brain infiltrating lymphocytes is critical for investigations involving murine neurological disease models. To optimize lymphocyte recovery, we compared two isolation methods using brains harvested from seven-day Theiler's murine encephalomyelitis virus (TMEV) and TMEV-OVA infected mice. Brains were processed using either a manual dounce based approach or enzymatic digestion using type IV collagenase. The resulting cell suspensions from these two techniques were transferred to a percoll gradient, centrifuged, and lymphocytes were recovered. Flow cytometric analysis of CD45hi cells showed greater percentage of CD44hiCD62lo activated lymphocytes and CD19+ B cells using the dounce method. In addition, we achieved a 3-fold greater recovery of activated virus-specific CD8 T cells specific for the immunodominant Db:VP2121-130 and engineered Kb:OVA257-264 epitopes through manual dounce homogenization approach as compared to collagenase digest. A greater percentage of viable cells was also achieved through dounce homogenization. Therefore, we conclude that manual homogenization is a superior approach to isolate activated T cells from the mouse brain.
Collapse
|
11
|
Qiao L, Zhang Y, Chai F, Tan Y, Huo C, Pan Z. Chimeric virus-like particles containing a conserved region of the G protein in combination with a single peptide of the M2 protein confer protection against respiratory syncytial virus infection. Antiviral Res 2016; 131:131-40. [PMID: 27154395 DOI: 10.1016/j.antiviral.2016.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 04/28/2016] [Accepted: 05/02/2016] [Indexed: 01/08/2023]
Abstract
To investigate the feasibility and efficacy of a virus-like particle (VLP) vaccine composed of the conserved antigenic epitopes of respiratory syncytial virus (RSV), the chimeric RSV VLPs HBcΔ-tG and HBcΔ-tG/M282-90 were generated based on the truncated hepatitis B virus core protein (HBcΔ). HBcΔ-tG consisted of HBcΔ, the conserved region (aa 144-204) of the RSV G protein. HBcΔ-tG was combined with a single peptide (aa 82-90) of the M2 protein to generate HBcΔ-tG/M282-90. Immunization of mice with the HBcΔ-tG or HBcΔ-tG/M282-90 VLPs elicited RSV-specific IgG and neutralizing antibody production and conferred protection against RSV infection. Compared with HBcΔ-tG, HBcΔ-tG/M282-90 induced decreased Th2 cytokine production (IL-4 and IL-5), increased Th1 cytokine response (IFN-γ, TNF-α, and IL-2), and increased ratios of IgG2a/IgG1 antibodies, thereby relieving pulmonary pathology upon subsequent RSV infection. Our results demonstrated that chimeric HBcΔ-tG/M282-90 VLPs represented an effective RSV subunit vaccine candidate.
Collapse
Affiliation(s)
- Lei Qiao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yuan Zhang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Feng Chai
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yiluo Tan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Chunling Huo
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Zishu Pan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
12
|
A phase 1a, first-in-human, randomized study of a respiratory syncytial virus F protein vaccine with and without a toll-like receptor-4 agonist and stable emulsion adjuvant. Vaccine 2016; 34:2847-54. [DOI: 10.1016/j.vaccine.2016.04.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 03/31/2016] [Accepted: 04/01/2016] [Indexed: 01/01/2023]
|
13
|
Grundy S, Plumb J, Kaur M, Ray D, Singh D. Additive anti-inflammatory effects of corticosteroids and phosphodiesterase-4 inhibitors in COPD CD8 cells. Respir Res 2016; 17:9. [PMID: 26809346 PMCID: PMC4727404 DOI: 10.1186/s12931-016-0325-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 01/13/2016] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND CD8 lymphocytes play an important role in the pathogenesis of COPD. Corticosteroids and phosphodiesterase 4 (PDE4) inhibitors are anti-inflammatory drugs used for COPD treatment. Little is known of the combined effect of these drugs on COPD CD8 cells. We studied the effect of corticosteroid combined with PDE4 inhibitors on cytokine release form circulating and pulmonary CD8 cells, and on glucocorticoid (GR) nuclear translocation. METHODS The effect of dexamethasone alone and in combination with the PDE4 inhibitors roflumilast and GSK256066 on cytokine release from circulating and pulmonary CD8 cells was measured. The effect of the compounds on nuclear translocation of GR and cyclic AMP-responsive element-binding protein (CREB) was studied using immunofluorescence. RESULTS Dexamethasone inhibited cytokine release from COPD CD8 cells in a concentration dependent manner. PDE4 inhibitors enhanced this anti-inflammatory effect in an additive manner. PDE4 inhibitors did not increase corticosteroid induced GR nuclear translocation. PDE4 inhibitors, but not corticosteroid, increased phospho-CREB nuclear translocation. CONCLUSION The combination of corticosteroids and PDE4 inhibitors results in an additive anti-inflammatory effect in COPD CD8 cells. This enhanced anti-inflammatory effect could translate to important clinical benefits for patients with COPD.
Collapse
Affiliation(s)
- Seamus Grundy
- Centre for Respiratory Medicine and Allergy, Institute of Inflammation and Repair, Manchester Academic Health Science Centre, The University of Manchester and University Hospital of South Manchester, NHS Foundation Trust Southmoor Road, Manchester, M23 9LT, UK.
| | - Jonathan Plumb
- Centre for Respiratory Medicine and Allergy, Institute of Inflammation and Repair, Manchester Academic Health Science Centre, The University of Manchester and University Hospital of South Manchester, NHS Foundation Trust Southmoor Road, Manchester, M23 9LT, UK
| | - Manminder Kaur
- Centre for Respiratory Medicine and Allergy, Institute of Inflammation and Repair, Manchester Academic Health Science Centre, The University of Manchester and University Hospital of South Manchester, NHS Foundation Trust Southmoor Road, Manchester, M23 9LT, UK
| | - David Ray
- School of Medicine and Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Dave Singh
- Centre for Respiratory Medicine and Allergy, Institute of Inflammation and Repair, Manchester Academic Health Science Centre, The University of Manchester and University Hospital of South Manchester, NHS Foundation Trust Southmoor Road, Manchester, M23 9LT, UK
| |
Collapse
|
14
|
Xu ML, Kim HJ, Wi GR, Kim HJ. The effect of dietary bovine colostrum on respiratory syncytial virus infection and immune responses following the infection in the mouse. J Microbiol 2015; 53:661-6. [PMID: 26310306 DOI: 10.1007/s12275-015-5353-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 08/03/2015] [Accepted: 08/03/2015] [Indexed: 10/23/2022]
Abstract
Human respiratory syncytial virus (hRSV) is the most common cause of respiratory tract infection among young children because of immature T cell immunity of them against hRSV. CD8 T cells play a pivotal role in clearing hRSV and preventing subsequent infection. We examined the effects of dietary bovine colostrum on virus infection and CD8 T cell responses following hRSV infection in the mouse model. Mice received bovine colostrum for 14 days prior to hRSV challenge, and lung indexes (severity of symptom) and lung virus titers were analyzed. In addition, the activation of CD8 T cells in the bronchoalveolar lavage fluids (BALFs) of mice receiving bovine colostrum were compared with those in the BALFs of mice receiving phosphate-buffered saline (PBS) or ribavirin, post virus challenge. The severity of infection and lung virus titers were reduced in the mice receiving bovine colostrum, compared to those receiving PBS. Moreover CD8 T cell responses were selectively enhanced in the former. Our results suggest that dietary bovine colostrum exerts the effects to inhibit hRSV and ameliorate the symptom by hRSV infection, and enhances the CD8 T cell response during the hRSV infection.
Collapse
Affiliation(s)
- Mei Ling Xu
- Laboratory of Virology, College of Pharmacy, Chung-Ang University, Seoul, 156-756, Republic of Korea
| | | | | | | |
Collapse
|
15
|
|