114 results on '"Tchilian, E"'
Search Results
2. Unusual case presentations associated with the CD45 C77G polymorphism
- Author
-
Tchilian, E. Z., Gil, J., Navarro, M. L., Fernandez-Cruz, E., Chapel, H., Misbah, S., Ferry, B., Renz, H., Schwinzer, R., and Beverley, P. C. L.
- Published
- 2006
3. Altered CD45 expression in C77G carriers influences immune function and outcome of hepatitis C infection
- Author
-
Dawes, R, Hennig, B, Irving, W, Petrova, S, Boxall, S, Ward, V, Wallace, D, Macallan, D C, Thursz, M, Hill, A, Bodmer, W, Beverley, P C L, and Tchilian, E Z
- Published
- 2006
4. Immunogenicity and Protective Efficacy of Seasonal Human Live Attenuated Cold-Adapted Influenza Virus Vaccine in Pigs.
- Author
-
Holzer, B, Morgan, SB, Martini, V, Sharma, R, Clark, B, Chiu, C, Salguero, FJ, Tchilian, E, Holzer, B, Morgan, SB, Martini, V, Sharma, R, Clark, B, Chiu, C, Salguero, FJ, and Tchilian, E
- Abstract
Influenza A virus infection is a global health threat to livestock and humans, causing substantial mortality and morbidity. As both pigs and humans are readily infected with influenza viruses of similar subtype, the pig is a robust and appropriate model for investigating swine and human disease. We evaluated the efficacy of the human cold-adapted 2017-2018 quadrivalent seasonal LAIV in pigs against H1N1pdm09 challenge. LAIV immunized animals showed significantly reduced viral load in nasal swabs. There was limited replication of the H1N1 component of the vaccine in the nose, a limited response to H1N1 in the lung lymph nodes and a low H1N1 serum neutralizing titer. In contrast there was better replication of the H3N2 component of the LAIV, accompanied by a stronger response to H3N2 in the tracheobronchial lymph nodes (TBLN). Our data demonstrates that a single administration of human quadrivalent LAIV shows limited replication in the nose and induces detectable responses to the H1N1 and H3N2 components. These data suggest that pigs may be a useful model for assessing LAIV against influenza A viruses.
- Published
- 2019
5. Induction of influenza-specific local CD8 T-cells in the respiratory tract after aerosol delivery of vaccine antigen or virus in the Babraham inbred pig
- Author
-
Tungatt, K, Dolton, G, Morgan, S, Attaf, M, Fuller, A, Whalley, T, Hemmink, J, Porter, E, Szomolay, B, Montoya, M, Hammond, J, Miles, J, Cole, D, Townsend, A, Bailey, M, Rizkallah, P, Charleston, B, Tchilian, E, and Sewell, A
- Subjects
Male ,Models, Molecular ,RNA viruses ,Viral Diseases ,Swine ,Respiratory System ,Sus scrofa ,CD8-Positive T-Lymphocytes ,Epitopes ,White Blood Cells ,Animal Cells ,Pig Models ,Medicine and Health Sciences ,Inbreeding ,Biology (General) ,Antigens, Viral ,Pathology and laboratory medicine ,Swine Diseases ,Mammals ,Staining ,T Cells ,Vaccination ,H1N1 ,Eukaryota ,Cell Staining ,Animal Models ,Medical microbiology ,Infectious Diseases ,Experimental Organism Systems ,Influenza A virus ,Influenza Vaccines ,Host-Pathogen Interactions ,Models, Animal ,Vertebrates ,Viruses ,Female ,Cellular Types ,Pathogens ,Research Article ,QH301-705.5 ,Immune Cells ,Immunology ,Cytotoxic T cells ,Research and Analysis Methods ,Microbiology ,Orthomyxoviridae Infections ,Influenza, Human ,Animals ,Humans ,Influenza viruses ,Amino Acid Sequence ,Molecular Biology Techniques ,Molecular Biology ,Aerosols ,Blood Cells ,Histocompatibility Antigens Class I ,Organisms ,Viral pathogens ,Biology and Life Sciences ,Cell Biology ,RC581-607 ,Influenza ,Microbial pathogens ,Specimen Preparation and Treatment ,Amniotes ,Immunologic diseases. Allergy ,Cloning ,Orthomyxoviruses - Abstract
There is increasing evidence that induction of local immune responses is a key component of effective vaccines. For respiratory pathogens, for example tuberculosis and influenza, aerosol delivery is being actively explored as a method to administer vaccine antigens. Current animal models used to study respiratory pathogens suffer from anatomical disparity with humans. The pig is a natural and important host of influenza viruses and is physiologically more comparable to humans than other animal models in terms of size, respiratory tract biology and volume. It may also be an important vector in the birds to human infection cycle. A major drawback of the current pig model is the inability to analyze antigen-specific CD8+ T-cell responses, which are critical to respiratory immunity. Here we address this knowledge gap using an established in-bred pig model with a high degree of genetic identity between individuals, including the MHC (Swine Leukocyte Antigen (SLA)) locus. We developed a toolset that included long-term in vitro pig T-cell culture and cloning and identification of novel immunodominant influenza-derived T-cell epitopes. We also generated structures of the two SLA class I molecules found in these animals presenting the immunodominant epitopes. These structures allowed definition of the primary anchor points for epitopes in the SLA binding groove and established SLA binding motifs that were used to successfully predict other influenza-derived peptide sequences capable of stimulating T-cells. Peptide-SLA tetramers were constructed and used to track influenza-specific T-cells ex vivo in blood, the lungs and draining lymph nodes. Aerosol immunization with attenuated single cycle influenza viruses (S-FLU) induced large numbers of CD8+ T-cells specific for conserved NP peptides in the respiratory tract. Collectively, these data substantially increase the utility of pigs as an effective model for studying protective local cellular immunity against respiratory pathogens., Author summary Influenza virus infection in pigs represents a significant problem to industry and also carries substantial risks to human health. Pigs can be infected with both bird and human forms of influenza where these viruses can mix with swine influenza viruses to generate new pandemic strains that can spread quickly and kill many millions of people across the globe. To date, the study of immunology and vaccination against flu in pigs has been hampered by a lack of suitable tools and reagents. Here, we have built a complete molecular toolset that allows such study. These tools could also be applied to other important infections in pigs such as foot-and-mouth disease and the normally fatal African Swine Fever virus. Finally, pigs are set to become an important model organism for study of influenza A virus infection. Here, we make use of a new research toolset to study a Broadly Protective Influenza Vaccine (BPIV) candidate, S-FLU, which could offer protection against all influenza A viruses. These new tools have been used to demonstrate the induction of large numbers of antigen specific CD8+ T cells to conserved NP epitopes in the respiratory tract after aerosol immunization.
- Published
- 2018
- Full Text
- View/download PDF
6. Anti-α4 integrin antibody induces apoptosis in murine thymocytes and staphylococcal enterotoxin B-activated lymph node T cells
- Author
-
TCHILIAN, E. Z., OWEN, J. J. T., and JENKINSON, E. J.
- Published
- 1997
7. Involvement of LFA-1/ICAM-2 adhesive interactions and PKC in activation-induced cell death following SEB rechallenge
- Author
-
TCHILIAN, E. Z., ANDERSON, G., MOORE, N. C., OWEN, J. J. T., and JENKINSON, E. J.
- Published
- 1996
8. The major histocompatibility complex homozygous inbred Babraham pig as a resource for veterinary and translational medicine
- Author
-
Schwartz, J. C., primary, Hemmink, J. D., additional, Graham, S. P., additional, Tchilian, E., additional, Charleston, B., additional, Hammer, S. E., additional, Ho, C.-S., additional, and Hammond, J. A., additional
- Published
- 2018
- Full Text
- View/download PDF
9. Targeted killing of colorectal cell lines and murine adenomas using a monoclonal antibody against membrane bound carcinoembryonic antigen
- Author
-
Conaghan, PJ, Wilding, JL, Tytherleigh, MG, Tchilian, E, Mortensen, NJM, and Bodmer, WF
- Published
- 2016
10. Murine CD33: Genomic structure, regulatory elements and splice variants
- Author
-
Watt, SM, LeeProudhoe, JE, Stubbs, L, Benton, MA, Ward, C, Hughes, J, Popov, B, and Tchilian, E
- Published
- 2016
11. Protein tyrosine phosphatase receptor type C polypeptide (PTPRC) on human chromosome band 1q31-->q32 localizes with marker D1S413(1) on a 610-kb yeast artificial chromosome
- Author
-
Goff, L, van Soest, S, Timón, M, Tchilian, E, and Beverley, P
- Published
- 2016
- Full Text
- View/download PDF
12. ESX1-dependent fractalkine mediates chemotaxis and Mycobacterium tuberculosis infection in humans
- Author
-
Hingley-Wilson, SM, Connell, D, Pollock, K, Hsu, T, Tchilian, E, Sykes, A, Grass, L, Potiphar, L, Bremang, S, Kon, OM, Jacobs, WR, Lalvani, A, and Medical Research Council (MRC)
- Subjects
EXPRESSION ,Microbiology (medical) ,ESX-1 ,Respiratory System ,Immunology ,Microbiology ,Antigens, CD11 ,Monocytes ,Fractalkine ,CX3CL1 ,Mycobacterium ,Bacterial Proteins ,Animals ,Humans ,Tuberculosis ,Cells, Cultured ,Mice, Inbred BALB C ,Science & Technology ,GRANULOMA-FORMATION ,CD11 Antigens ,Chemokine CX3CL1 ,INDUCTION ,Chemotaxis ,Macrophages ,ESAT-6/CFP-10 ,11 Medical And Health Sciences ,Mycobacterium tuberculosis ,CHEMOKINE ,Matrix Metalloproteinases ,Mechanisms of Pathogenesis ,Infectious Diseases ,CALMETTE-GUERIN ,SURVIVAL ,VIRULENCE ,Infection ,Life Sciences & Biomedicine ,RC - Abstract
Summary Mycobacterium tuberculosis-induced cellular aggregation is essential for granuloma formation and may assist establishment and early spread of M. tuberculosis infection. The M. tuberculosis ESX1 mutant, which has a non-functional type VII secretion system, induced significantly less production of the host macrophage-derived chemokine fractalkine (CX3CL1). Upon infection of human macrophages ESX1-dependent fractalkine production mediated selective recruitment of CD11b+ monocytic cells and increased infection of neighbouring cells consistent with early local spread of infection. Fractalkine levels were raised in vivo at tuberculous disease sites in humans and were significantly associated with increased CD11b+ monocytic cellular recruitment and extent of granulomatous disease. These findings suggest a novel fractalkine-dependent ESX1-mediated mechanism in early tuberculous disease pathogenesis in humans. Modulation of M. tuberculosis-mediated fractalkine induction may represent a potential treatment option in the future, perhaps allowing us to switch off a key mechanism required by the pathogen to spread between cells.
- Published
- 2014
13. Immunogenicity and protective efficacy of prime-boost regimens with recombinant (delta)ureC hly+ Mycobacterium bovis BCG and modified vaccinia virus ankara expressing M. tuberculosis antigen 85A against murine tuberculosis
- Author
-
Tchilian, E, Desel, C, Forbes, E, Bandermann, S, Sander, C, Hill, A, Mcshane, H, and Kaufmann, S
- Subjects
complex mixtures - Abstract
In the light of the recent emergence of multidrug-resistant and extensively drug-resistant strains of Mycobacterium tuberculosis, the epidemic of tuberculosis (TB) in populations coinfected with human immunodeficiency virus, and the failure of Mycobacterium bovis bacillus Calmette-Guerin (BCG) to protect against disease, new vaccines against TB are urgently needed. Two promising new vaccine candidates are the recombinant DeltaureC hly(+) BCG (recBCG), which has been developed to replace the current BCG vaccine strain, and modified vaccinia virus Ankara (MVA) expressing M. tuberculosis antigen 85A (MVA85A), which is a leading candidate vaccine designed to boost the protective efficacy of BCG. In the present study, we examined the effect of MVA85A boosting on the protection afforded at 12 weeks postchallenge by BCG and recBCG by using bacterial CFU as an efficacy readout. recBCG-immunized mice were significantly better protected against aerosol challenge with M. tuberculosis than mice immunized with the parental strain of BCG. Intradermal boosting with MVA85A did not reduce the bacterial burden any further. In order to identify a marker for the development of a protective immune response against M. tuberculosis challenge, we analyzed splenocytes after priming or prime-boosting by using intracytoplasmic cytokine staining and assays for cytokine secretion. Boosting with MVA85A, but not priming with BCG or recBCG, greatly increased the antigen 85A-specific T-cell response, suggesting that the mechanism of protection may differ from that against BCG or recBCG. We show that the numbers of systemic multifunctional cytokine-producing cells did not correlate with protection against aerosol challenge in BALB/c mice. This emphasizes the need for new biomarkers for the evaluation of TB vaccine efficacy.
- Published
- 2009
14. New tuberculosis vaccines can diminish as well as enhance BCG induced protection: Results from a murine model
- Author
-
Sander, C, Tchilian, E, Dahm-Vicker, M, Fletcher, H, Mcshane, H, and Hill, A
- Published
- 2006
15. Harnessing local and systemic immunity for vaccines against tuberculosis
- Author
-
Beverley, P C L, primary, Sridhar, S, additional, Lalvani, A, additional, and Tchilian, E Z, additional
- Published
- 2014
- Full Text
- View/download PDF
16. A point mutation in CD45 may be associated with an increased risk of HIV-1 infection (vol 15, pg 1892, 2001)
- Author
-
Tchilian, E, Wallace, D, Dawes, R, Imami, N, Burton, C, Gotch, F, and Beverly, P
- Published
- 2001
17. Erratum: A point mutation in CD45 may be associated with an increased risk of HIV-1 infection (AIDS (2001) 15 (1892-1894)). [2]
- Author
-
Tchilian, E, Wallace, D, Dawes, R, Imami, N, Burton, C, Gotch, F, and Beverly, P
- Published
- 2001
18. Targeted killing of colorectal cancer cell lines by a humanised IgG1 monoclonal antibody that binds to membrane-bound carcinoembryonic antigen
- Author
-
Conaghan, P J, primary, Ashraf, S Q, additional, Tytherleigh, M G, additional, Wilding, J L, additional, Tchilian, E, additional, Bicknell, D, additional, Mortensen, N JMcC, additional, and Bodmer, W F, additional
- Published
- 2008
- Full Text
- View/download PDF
19. Altered CD45 isoform expression affects lymphocyte function in CD45 Tg mice
- Author
-
Tchilian, E. Z., primary
- Published
- 2004
- Full Text
- View/download PDF
20. Protein tyrosine phosphatase receptor type C polypeptide (PTPRC) on human chromosome band 1q31→q32 localizes with marker D1S4131 on a 610-kb yeast artificial chromosome
- Author
-
Goff, L.K., primary, van Soest, S., additional, Timón, M., additional, Tchilian, E., additional, and Beverley, P.C.L., additional
- Published
- 1999
- Full Text
- View/download PDF
21. CD66 identifies a neutrophil-specific epitope within the hematopoietic system that is expressed by members of the carcinoembryonic antigen family of adhesion molecules
- Author
-
Watt, SM, primary, Sala-Newby, G, additional, Hoang, T, additional, Gilmore, DJ, additional, Grunert, F, additional, Nagel, G, additional, Murdoch, SJ, additional, Tchilian, E, additional, Lennox, ES, additional, and Waldmann, H, additional
- Published
- 1991
- Full Text
- View/download PDF
22. Protein tyrosine phosphatase receptor type C polypeptide (PTPRC) on human chromosome band 1q31→q32 localizes with marker D1S413<FOOTREF>[sup 1] </FOOTREF> on a 610-kb yeast artificial chromosome.
- Author
-
Goff, L.K., Soest, S. Van, Timón, M., Tchilian, E., and Beverley, P.C.L.
- Subjects
GENE mapping ,HUMAN chromosomes ,ARTIFICIAL chromosomes ,DNA ,LINKAGE (Genetics) ,GENETIC markers ,PULSED-field gel electrophoresis - Abstract
This article reports that the 120-kb Protein tyrosine phosphatase receptor type C polypeptide (PTPRC) gene co-localizes with marker D1S413 on a 610-kb yeast artificial chromosome (YAC) and therefore considerably refines the previous physical localization of this gene on chromosome 1q31→q32. High molecular weight DNA was prepared in high concentration agarose plugs. The size of the YAC was determined by Pulsed Field Gel Electrophoresis using 1 % PFGE grade agarose with a CHEF DRII system run for 24 h on a 30-70 s pulse ramp, 6 V/cm at 10-15°C in 0.5 x TBE, blotted and then hybridised with PTPRC 5' probe. Linkage analysis has mapped the calcium channel &b.alpha;1 subunit gene, CACNL1A3, to a 5-cM interval at 1q31→q32 shared by D1S413 and has revealed that CACNL1A3 lies telomeric to D1S413.
- Published
- 1999
- Full Text
- View/download PDF
23. Protein tyrosine phosphatase receptor type C polypeptide (PTPRC) on human chromosome band 1q31→q32 localizes with marker D1S413<footref rid="foot01">1</footref> on a 610-kb yeast artificial chromosome.
- Author
-
Goff, L.K., van Soest, S., Timón, M., Tchilian, E., and Beverley, P.C.L.
- Published
- 1999
- Full Text
- View/download PDF
24. Respiratory immunisation and mucosal immunity against influenza
- Author
-
Martini, V, Tchilian, E, Townsend, A, Drakesmith, A, Graham, S, and MacLoughlin, R
- Subjects
Influenza vaccines ,Immunology - Abstract
Influenza A virus (IAV) is a considerable threat to human and animal health. Current immunisation strategies rely primarily on inducing strain-specific neutralising antibodies with a limited breadth of protection. Increasing evidence indicates the importance of local immune responses and tissue resident memory T cells (TRM) for broad protection against respiratory infections, however, a vaccine that can be safely delivered to the lung is yet to be approved. In addition, there is little information on influenza A virus (IAV) specific TRM in the pig, a large animal model for IAV research, and a natural reservoir for the virus. The first part of this thesis describes the characterisation of porcine CD8 TRM in the lungs and upper respiratory tract following H1N1pmd09 infection. The phenotype of CD8 T cells specific for three influenza nucleoprotein (NP) epitopes, detected using MHC class I tetramers, was determined. This analysis revealed that the hierarchy of response differed with time in different tissues. The frequency of NP-specific cells declined over 63 days in all tissues, but it was best maintained in the lung. Most tetramer+ CD8 T cells isolated from broncho-alveolar lavage (BAL) displayed a TRM phenotype, expressing CD69 and an effector memory phenotype. NP-specific T cells isolated from BAL presented genes characteristic of human TRM, but gene expression differed at 7, 21 and 63 days post-infection. It is essential to assess vaccine deposition to provide optimal delivery of vaccines to the respiratory tract. The distribution of droplets generated by a commonly used intranasal device (MAD) and two commercially available vibrating mesh nebulizers (VMNs) was therefore compared. Droplets generated with the drug albuterol or a radiolabel (99mTc-DTPA), or a model vaccine (S-FLU) presented similar aerosol characteristics. A scintigraphic study using 99mTc-DTPA showed that VMNs uniformly distributed the radiolabel in the porcine lung and upper respiratory tract. The intranasal administration of a large volume (1ml/nostril) by MAD also delivered a high proportion of the dose to the lungs, although in a concentrated and restricted area. Recombinant single-cycle IAV viruses, such as S-FLU, may offer a promising solution to currently available vaccines as they can be delivered safely to the lung and elicit heterotypic immunity. However, previous studies in pigs showed a limited reduction in viral replication following challenge. In the last chapter, two strategies to improve S-FLU immunogenicity and protection are presented: 1) implementing a second-generation version of S-FLU expressing HA (CLEARFLU) and 2) administering S-FLU simultaneously by aerosol and intramuscular injection (SIM). CLEARFLU did not generate neutralising antibodies, however, S-FLU administered by SIM produced a high titer of neutralising antibody, robust local cellular responses and conferred protection against homologous challenge. On the other hand, intramuscular injection alone elicited humoral responses in the periphery, but failed to do so locally, and did not stimulate cellular responses. Aerosol S-FLU did not protect despite high T cell responses in the respiratory tract, probably due to the lack of neutralising antibody in the periphery. These data showed for the first time that porcine TRM present similar properties to those of humans, further establishing the pig as a useful model for investigation of local memory responses following respiratory infection. In addition, aerosol delivery can uniformly target the lungs but only the SIM immunisation regime successfully protected pigs from challenge, warranting further studies of this immunisation strategy against IAV and other respiratory pathogens.
- Published
- 2022
25. Evaluation of a single cycle influenza virus as a candidate vaccine
- Author
-
Sadler, H, Tchilian, E, and Townsend, A
- Subjects
Influenza vaccines - Abstract
Influenza A continues to impose a considerable burden on humans and animals, including farmed poultry and pigs. Current vaccination approaches successfully protect certain populations against specific strains, but none are effective at consistently inducing broad and long-lasting protection to the diversity of circulating strains. Single cycle viruses may offer a promising solution as they are able to safely present a wide array of viral antigens in the respiratory tract by undergoing just one round of cell infection in their host. This is thought to be advantageous for stimulating broadly protective resident memory T-cell responses in the lung as well as mucosal antibody responses. This thesis describes the design and evaluation of a candidate vaccine platform called CLEARFLU, building on the previously described single cycle virus S-Flu designed by Alain Townsend. CLEARFLU viruses are unable to replicate in vivo due to the introduction of several independently inactivating mutations into the hemagglutinin protein, blocking the cell entry step of infection. They can however be propagated in cell culture by providing complementing hemagglutinin in trans. We demonstrate that CLEARFLU viruses do not cause disease in mice or pigs, even when administered to the respiratory tract. They induce a strong T-cell response in the lung, including to the highly conserved viral component nucleoprotein which is known to be associated with broad protection. CLEARFLU viruses also stimulate both systemic and local strain-specific antibody to neuraminidase and neutralising antibody to hemagglutinin in mice. This neutralising response to hemagglutinin is a notable advantage over other hemagglutinin-inactivated single cycle viruses like S-Flu which do not induce such responses and can thus limit but not prevent future infection. Together with investigations into their genomic stability, we demonstrate that CLEARFLU viruses show promise as vaccine candidates able to generate both non-neutralising protection against a broad range of strains and neutralising protection against strain-matched challenge.
- Published
- 2021
26. Mucosal T cell defence in large animal models of infection
- Author
-
Edmans, M, Benedictus, L, Sidonia, E, Klenerman, P, Tchilian, E, and Christopher, N
- Subjects
Immunology, Comparative ,Immunology ,Influenza - Abstract
The study of cellular immune responses in the mucosa has proven difficult within humans. Capturing the respiratory mucosal immune response is challenging without invasive sampling methods; therefore, research in this area has leant heavily on the use of animal models. Livestock species are naturally infected by a range of diseases which are also major human public health concerns, including influenza in pigs, tuberculosis in cattle and lung disease in sheep. Influenza virus is estimated to cause 1 billion human infections annually with approximately 3- 5 million cases of severe illness and 300-500,000 deaths per year. Pigs make an ideal model for the study of influenza infection as they have comparable size and respiratory anatomy to humans and influenza strains such as the 2009 pandemic H1N1 are endemic in both humans and pigs. The T cell response to influenza has come under renewed investigation in recent years with the potential to target cross reactive T cell epitopes to multiple influenza strains. Additionally, innate like T cells, such as mucosal associated invariant T (MAIT) cells, a prominent T cell population in humans, are being increasingly studied for their role in influenza infection. Here, the kinetic of virus infection and T cell response to H1N1pdm09 influenza is investigated in inbred Babraham pigs and compared to commercial outbred animals. High level of nasal virus shedding continued up to day 4-5 post infection followed by a steep decline and clearance of virus by day 9. The T cell response developed from day 5-6 post infection reaching a peak at 9-14 days. BAL contained the most highly activated CD8 and CD4 T cells actively producing pro-inflammatory cytokines, which likely contribute to the elimination of the virus. The weak response in peripheral blood T cells did not reflect the robust local immune responses. The immune responses in the Babraham pig were comparable to outbred pigs, validating the Babraham pig as a model for pig immunology. Furthermore, new H1N1pdm09 influenza immunodominant T cell epitopes are described in the Babraham pig which will aid in future studies involving this animal model. Putative MAIT cells have been previously identified in pigs, cattle and sheep by qPCR-based methods though no MAIT cells have been phenotypically described in these species. In this study the MAIT cell population in pigs, cattle and sheep is determined using a variety of methods including functional activation assays and phenotypic assessment utilising species specific and xenogenic tetramers. In pigs the MAIT cell population was at the limits of detection. The absence of MAIT cells identified in pigs precluded the study of MAIT cells in the context of the influenza challenge studies. However, MAIT cells were identified in sheep and cattle and a detailed characterization was therefore performed in cattle. MAIT cells in cattle were highly phenotypically and functionally comparable to MAIT cells described in humans and mice and are activated by disease causing bacteria in vitro and in vivo. These data and techniques expand the understanding of the T cell response to influenza in pigs, support the use of the Babraham pig as a model for influenza research and enables cattle and sheep to be included as useful models for the study of MAIT cells in these species.
- Published
- 2021
27. Understanding vaccine induced protective immunity to Mycobacterium tuberculosis
- Author
-
Ronan, E, Beverley, P, Tchilian, E, and Sattentau, Q
- Subjects
Vaccinology ,Immunology ,Biology (medical sciences) ,Infectious diseases - Abstract
The current worldwide epidemic of Mycobacterium tuberculosis infection is a huge global health problem. Widespread BCG vaccination remains a useful tool in combating this epidemic; however, its variable efficacy requires urgent development of novel vaccines against Mycobacterium tuberculosis. Such a candidate vaccine is a serotype 5 adenovirus expressing antigen 85A from M. tuberculosis (Ad85A). In animal models Ad85A confers significant protection when administered intra-nasally. The work in this thesis demonstrates that intra-nasal immunisation with Ad85A results in inhibition of M. tuberculosis growth in the lung early after infection, in contrast to the late inhibition induced by parenterally administered vaccines. Early inhibition correlates with the presence in the lung of a highly activated population of antigen-specific CD8 T cells, maintained for at least 6 months post-immunisation by persistent antigen. For intra-nasal Ad85A to be effective, the vaccine must be delivered into the lower respiratory tract, as immunisation targeting only the nasal-associated lymphoid tissue (NALT) does not result in protection. Following a change of animal facility, the lung immune response to intra-dermal immunisation with Ad85A increased and this route of immunisation now induced protection, though growth of M. tuberculosis was inhibited only late after infection. However, this response and protection can be altered by exposure to environmental mycobacteria. Further experiments showed that simultaneous respiratory and parenteral immunisations (SIM) act additively, where local lung immunity inhibits the growth of M. tuberculosis early after infection and systemic immunity protects later. SIM regimes generate greatly improved protection over either immunisation alone and do not depend on priming and boosting.
- Published
- 2016
28. Development of bovine IgG3-specific assays using a novel recombinant single-domain binding reagent.
- Author
-
Noble A, Moorhouse E, Hay AL, Paudyal B, Mwangi W, Munir D, Placido MB, Tchilian E, Hammond JA, and Graham SP
- Subjects
- Animals, Cattle, Antibodies, Viral blood, Antibodies, Viral immunology, Ankyrin Repeat, Immunoglobulin G blood, Immunoglobulin G immunology, Enzyme-Linked Immunosorbent Assay veterinary, Recombinant Proteins immunology, Antibodies, Monoclonal immunology
- Abstract
Cattle express three subclasses of IgG antibody - IgG1, IgG2 and IgG3. Unlike IgG1 and IgG2, IgG3 was described relatively recently and the role of this subclass in immunity is unknown. Using recombinant bovine IgG1, IgG2 and IgG3 monoclonal antibodies (mAbs), we demonstrated that only one of the commercially available anti-bovine IgG mAbs tested was able to recognize IgG3, and no mAb exclusively bound IgG3. Here, we evaluated a small ankyrin repeat protein, called Ankyron™ AZS40101, that was generated to bind to bovine IgG3. Ankyron™ AZS40101 bound specifically to IgG3 with minimal reactivity to IgG1 and IgG2. Ankyron™ AZS40101 was shown to be useful in ELISA assays as either a capture or detection reagent. Utilisation of Ankyron™ AZS40101 alongside IgG1 and IgG2 specific mAbs to detect antigen-specific IgG subclasses in the serum of cattle sequentially vaccinated with heterologous foot-and-mouth disease virus capsid antigens revealed a low-level antigen-specific IgG3 response, in addition to IgG1 and IgG2 responses. Assessment of the total IgG1, IgG2 and IgG3 levels in healthy cattle plasma samples showed that IgG3 was measurable at a mean concentration of 0.19 mg/mL, although this was significantly lower than those of IgG1 (mean 3.28 mg/mL) and IgG2 (mean 3.41 mg/mL). Thus, Ankyron™ AZS40101 is a new reagent that provides utility for measurement of bovine IgG3 responses to infection and vaccination., Competing Interests: Declaration of Competing Interest None, (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
29. Comparative pharmacokinetics of porcine and human anti-influenza hemagglutinin monoclonal antibodies in outbred pigs and minipigs.
- Author
-
Paudyal B, Moorhouse E, Sharma B, Dodds M, Nguyen V, Milad M, and Tchilian E
- Subjects
- Animals, Swine, Humans, Orthomyxoviridae Infections immunology, Half-Life, Female, Influenza, Human immunology, Swine, Miniature, Antibodies, Monoclonal pharmacokinetics, Hemagglutinin Glycoproteins, Influenza Virus immunology, Antibodies, Viral immunology
- Abstract
Assessing the pharmacokinetics of monoclonal antibodies (mAbs) in relevant animal models is essential for designing improved formulations and developing mAb delivery platforms. We have established the pig, a large natural host animal for influenza with many similarities to humans, as a robust model for testing the therapeutic efficacy of anti-influenza mAbs and evaluating mAb delivery platforms. Here, we compared the pharmacokinetic characteristics of two anti-influenza hemagglutinin mAbs, human 2-12C and porcine pb27, in Göttingen minipigs and Landrace × Large White outbred pigs. Minipigs offer the advantage of a more stable weight, whereas outbred pigs are more readily available but exhibit rapid growth. Outbred pigs and minipigs showed similar pharmacokinetics and a similar porcine pb27 half-life (half-life of 15.7 days for outbred pigs and 16.6 days for minipigs). In contrast, the half-life of human 2-12C was more rapid in two of the minipigs but not in the outbred pigs, correlating with the development of antidrug antibodies in the two minipigs. Our results demonstrate that both outbred pigs and minipigs are appropriate models for pharmacokinetic studies and the evaluation of mAb delivery platforms, potentially bridging the gap between small animals and human trials., Competing Interests: MD was employed by the company Certara. VN and MM were employed by the company Milad Pharmaceutical Consulting LLC. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Paudyal, Moorhouse, Sharma, Dodds, Nguyen, Milad and Tchilian.)
- Published
- 2024
- Full Text
- View/download PDF
30. Aerosol immunization with influenza matrix, nucleoprotein, or both prevents lung disease in pig.
- Author
-
Vatzia E, Paudyal B, Dema B, Carr BV, Sedaghat-Rostami E, Gubbins S, Sharma B, Moorhouse E, Morris S, Ulaszewska M, MacLoughlin R, Salguero FJ, Gilbert SC, and Tchilian E
- Abstract
Current influenza vaccines are strain-specific and require frequent updates to combat new strains, making a broadly protective influenza vaccine (BPIV) highly desirable. A promising strategy is to induce T-cell responses against internal proteins conserved across influenza strains. In this study, pH1N1 pre-exposed pigs were immunized by aerosol using viral vectored vaccines (ChAdOx2 and MVA) expressing matrix (M1) and nucleoprotein (NP). Following H3N2 challenge, all immunizations (M1, NP or NPM1) reduced lung pathology, but M1 alone offered the greatest protection. NP or NPM1 immunization induced both T-cell and antibody responses. M1 immunization generated no detectable antibodies but elicited M1-specific T-cell responses, suggesting T cell-mediated protection. Additionally, a single aerosol immunization with the ChAdOx vaccine encoding M1, NP and neuraminidase reduced lung pathology. These findings provide insights into BPIV development using a relevant large natural host, the pig., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
31. Single-cell analysis reveals lasting immunological consequences of influenza infection and respiratory immunization in the pig lung.
- Author
-
Muir A, Paudyal B, Schmidt S, Sedaghat-Rostami E, Chakravarti S, Villanueva-Hernández S, Moffat K, Polo N, Angelopoulos N, Schmidt A, Tenbusch M, Freimanis G, Gerner W, Richard AC, and Tchilian E
- Subjects
- Animals, Swine, Immunization, Bronchoalveolar Lavage Fluid immunology, Bronchoalveolar Lavage Fluid virology, Single-Cell Analysis, Orthomyxoviridae Infections immunology, Orthomyxoviridae Infections virology, Lung immunology, Lung virology, Influenza Vaccines immunology, Influenza A Virus, H1N1 Subtype immunology
- Abstract
The pig is a natural host for influenza viruses and integrally involved in virus evolution through interspecies transmissions between humans and swine. Swine have many physiological, anatomical, and immunological similarities to humans, and are an excellent model for human influenza. Here, we employed single cell RNA-sequencing (scRNA-seq) and flow cytometry to characterize the major leukocyte subsets in bronchoalveolar lavage (BAL), twenty-one days after H1N1pdm09 infection or respiratory immunization with an adenoviral vector vaccine expressing hemagglutinin and nucleoprotein with or without IL-1β. Mapping scRNA-seq clusters from BAL onto those previously described in peripheral blood facilitated annotation and highlighted differences between tissue resident and circulating immune cells. ScRNA-seq data and functional assays revealed lasting impacts of immune challenge on BAL populations. First, mucosal administration of IL-1β reduced the number of functionally active Treg cells. Second, influenza infection upregulated IFI6 in BAL cells and decreased their susceptibility to virus replication in vitro. Our data provide a reference map of porcine BAL cells and reveal lasting immunological consequences of influenza infection and respiratory immunization in a highly relevant large animal model for respiratory virus infection., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2024 Muir et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2024
- Full Text
- View/download PDF
32. MAIT cell-MR1 reactivity is highly conserved across multiple divergent species.
- Author
-
Edmans MD, Connelley TK, Morgan S, Pediongco TJ, Jayaraman S, Juno JA, Meehan BS, Dewar PM, Maze EA, Roos EO, Paudyal B, Mak JYW, Liu L, Fairlie DP, Wang H, Corbett AJ, McCluskey J, Benedictus L, Tchilian E, Klenerman P, and Eckle SBG
- Subjects
- Animals, Humans, Mice, Cattle, Swine, Macaca, Receptors, Antigen, T-Cell, alpha-beta immunology, Receptors, Antigen, T-Cell, alpha-beta metabolism, Receptors, Antigen, T-Cell, alpha-beta genetics, Mucosal-Associated Invariant T Cells immunology, Mucosal-Associated Invariant T Cells metabolism, Histocompatibility Antigens Class I immunology, Histocompatibility Antigens Class I metabolism, Minor Histocompatibility Antigens metabolism, Minor Histocompatibility Antigens genetics, Minor Histocompatibility Antigens immunology, Minor Histocompatibility Antigens chemistry, Species Specificity
- Abstract
Mucosal-associated invariant T (MAIT) cells are a subset of unconventional T cells that recognize small molecule metabolites presented by major histocompatibility complex class I related protein 1 (MR1), via an αβ T cell receptor (TCR). MAIT TCRs feature an essentially invariant TCR α-chain, which is highly conserved between mammals. Similarly, MR1 is the most highly conserved major histocompatibility complex-I-like molecule. This extreme conservation, including the mode of interaction between the MAIT TCR and MR1, has been shown to allow for species-mismatched reactivities unique in T cell biology, thereby allowing the use of selected species-mismatched MR1-antigen (MR1-Ag) tetramers in comparative immunology studies. However, the pattern of cross-reactivity of species-mismatched MR1-Ag tetramers in identifying MAIT cells in diverse species has not been formally assessed. We developed novel cattle and pig MR1-Ag tetramers and utilized these alongside previously developed human, mouse, and pig-tailed macaque MR1-Ag tetramers to characterize cross-species tetramer reactivities. MR1-Ag tetramers from each species identified T cell populations in distantly related species with specificity that was comparable to species-matched MR1-Ag tetramers. However, there were subtle differences in staining characteristics with practical implications for the accurate identification of MAIT cells. Pig MR1 is sufficiently conserved across species that pig MR1-Ag tetramers identified MAIT cells from the other species. However, MAIT cells in pigs were at the limits of phenotypic detection. In the absence of sheep MR1-Ag tetramers, a MAIT cell population in sheep blood was identified phenotypically, utilizing species-mismatched MR1-Ag tetramers. Collectively, our results validate the use and define the limitations of species-mismatched MR1-Ag tetramers in comparative immunology studies., Competing Interests: Conflict of interest J. Y. W. M., L. L., D. P. F., A. J. C., J. M., and S. B. G. E. are inventors on university owned patent rights (patent families WO/2015/149130 and WO/2014/005194) licensed for commercial use to Immudex and for non-profit use to the NIH Tetramer Core Facility. All other authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
33. Distinct effector functions mediated by Fc regions of bovine IgG subclasses and their interaction with Fc gamma receptors.
- Author
-
Noble A, Paudyal B, Schwartz JC, Mwangi W, Munir D, Tchilian E, Hammond JA, and Graham SP
- Subjects
- Humans, Cattle, Animals, Mice, Swine, Immunologic Factors, Macrophages, Phagocytosis, Antibodies, Monoclonal, Antigens, Receptors, IgG, Immunoglobulin G
- Abstract
Cattle possess three IgG subclasses. However, the key immune functions, including complement and NK cell activation, and enhancement of phagocytosis, are not fully described for bovine IgG1, 2 and 3. We produced chimeric monoclonal antibodies (mAbs) consisting of a defined variable region linked to the constant regions of bovine IgG1, 2 and 3, and expressed His-tagged soluble recombinant bovine Fc gamma receptors (FcγRs) IA (CD64), IIA (CD32A), III (CD16) and Fcγ2R. Functional assays using bovinized mAbs were developed. IgG1 and IgG3, but not IgG2, activated complement-dependent cytotoxicity. Only IgG1 could activate cattle NK cells to mobilize CD107a after antigen crosslinking, a surrogate assay for antibody-dependent cell cytotoxicity. Both IgG1 and IgG2 could trigger monocyte-derived macrophages to phagocytose fluorescently labelled antigen-expressing target cells. IgG3 induced only weak antibody-dependent cellular phagocytosis (ADCP). By contrast, monocytes only exhibited strong ADCP when triggered by IgG2. IgG1 bound most strongly to recombinant FcγRs IA, IIA and III, with weaker binding by IgG3 and none by IgG2, which bound exclusively to Fcγ2R. Immune complexes containing IgG1, 2 and 3 bound differentially to leukocyte subsets, with IgG2 binding strongly to neutrophils and monocytes and all subclasses binding platelets. Differential expression of the FcγRs on leukocyte subsets was demonstrated by surface staining and/or RT-qPCR of sorted cells, e.g., Fcγ2R mRNA was expressed in monocytes/macrophages, neutrophils, and platelets, potentially explaining their strong interactions with IgG2, and FcγRIII was expressed on NK cells, presumably mediating IgG1-dependent NK cell activation. These data reveal differences in bovine IgG subclass functionality, which do not correspond to those described in humans, mice or pigs, which is relevant to the study of these IgG subclasses in vaccine and therapeutic antibody development., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2023 Noble, Paudyal, Schwartz, Mwangi, Munir, Tchilian, Hammond and Graham.)
- Published
- 2023
- Full Text
- View/download PDF
34. A direct contact pig influenza challenge model for assessing protective efficacy of monoclonal antibodies.
- Author
-
McNee A, Vanover D, Rijal P, Paudyal B, Lean FZX, MacLoughlin R, Núñez A, Townsend A, Santangelo PJ, and Tchilian E
- Subjects
- Swine, Humans, Animals, Antibodies, Monoclonal, Respiratory Aerosols and Droplets, Hemagglutinins, Influenza, Human, Orthomyxoviridae Infections
- Abstract
Monoclonal antibodies (mAbs) can be used to complement immunization for the therapy of influenza virus infection. We have established the pig, a natural large animal host for influenza A, with many physiological, immunological, and anatomical similarities to humans, as an appropriate model for testing mAbs. We have evaluated the protective efficacy of the strongly neutralizing human anti-hemagglutinin mAb, 2-12C in the pig influenza model. Intravenous administration of recombinant 2-12C reduced virus load and lung pathology, however, it did not prevent virus nasal shedding and, consequently, transmission. This may be because the pigs were directly infected intranasally with a high dose of the H1N1pdm09 virus. To address this, we developed a contact challenge model in which the animals were given 2-12C and one day later co-housed with donor pigs previously infected intra-nasally with H1N1pdm09. 2-12C pre-treatment completely prevented infection. We also administered a lower dose of 2-12C by aerosol to the respiratory tract, but this did not prevent shedding in the direct challenge model, although it abolished lung infection. We propose that the direct contact challenge model of pig influenza may be useful for evaluating candidate mAbs and emerging delivery platforms prior to clinical trials., Competing Interests: Author RM is an employee of the company Aerogen Limited. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 McNee, Vanover, Rijal, Paudyal, Lean, MacLoughlin, Núñez, Townsend, Santangelo and Tchilian.)
- Published
- 2023
- Full Text
- View/download PDF
35. Simultaneous co-infection with swine influenza A and porcine reproductive and respiratory syndrome viruses potentiates adaptive immune responses.
- Author
-
Chrun T, Maze EA, Roper KJ, Vatzia E, Paudyal B, McNee A, Martini V, Manjegowda T, Freimanis G, Silesian A, Polo N, Clark B, Besell E, Booth G, Carr BV, Edmans M, Nunez A, Koonpaew S, Wanasen N, Graham SP, and Tchilian E
- Subjects
- Animals, Swine, Humans, Influenza A Virus, H3N2 Subtype, Immunity, Porcine respiratory and reproductive syndrome virus, Porcine Reproductive and Respiratory Syndrome, Coinfection, Influenza, Human
- Abstract
Porcine respiratory disease is multifactorial and most commonly involves pathogen co-infections. Major contributors include swine influenza A (swIAV) and porcine reproductive and respiratory syndrome (PRRSV) viruses. Experimental co-infection studies with these two viruses have shown that clinical outcomes can be exacerbated, but how innate and adaptive immune responses contribute to pathogenesis and pathogen control has not been thoroughly evaluated. We investigated immune responses following experimental simultaneous co-infection of pigs with swIAV H3N2 and PRRSV-2. Our results indicated that clinical disease was not significantly exacerbated, and swIAV H3N2 viral load was reduced in the lung of the co-infected animals. PRRSV-2/swIAV H3N2 co-infection did not impair the development of virus-specific adaptive immune responses. swIAV H3N2-specific IgG serum titers and PRRSV-2-specific CD8β
+ T-cell responses in blood were enhanced. Higher proportions of polyfunctional CD8β+ T-cell subset in both blood and lung washes were found in PRRSV-2/swIAV H3N2 co-infected animals compared to the single-infected groups. Our findings provide evidence that systemic and local host immune responses are not negatively affected by simultaneous swIAV H3N2/PRRSV-2 co-infection, raising questions as to the mechanisms involved in disease modulation., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Chrun, Maze, Roper, Vatzia, Paudyal, McNee, Martini, Manjegowda, Freimanis, Silesian, Polo, Clark, Besell, Booth, Carr, Edmans, Nunez, Koonpaew, Wanasen, Graham and Tchilian.)- Published
- 2023
- Full Text
- View/download PDF
36. Effect of mucosal adjuvant IL-1β on heterotypic immunity in a pig influenza model.
- Author
-
Schmidt A, Paudyal B, Villanueva-Hernández S, Mcnee A, Vatzia E, Carr BV, Schmidt S, Mccarron A, Martini V, Schroedel S, Thirion C, Waters R, Salguero FJ, Gerner W, Tenbusch M, and Tchilian E
- Subjects
- Animals, Humans, Adjuvants, Immunologic, Antibodies, Viral, Influenza A Virus, H3N2 Subtype, Swine, Influenza A Virus, H1N1 Subtype, Orthomyxoviridae Infections
- Abstract
T cell responses directed against highly conserved viral proteins contribute to the clearance of the influenza virus and confer broadly cross-reactive and protective immune responses against a range of influenza viruses in mice and ferrets. We examined the protective efficacy of mucosal delivery of adenoviral vectors expressing hemagglutinin (HA) and nucleoprotein (NP) from the H1N1 virus against heterologous H3N2 challenge in pigs. We also evaluated the effect of mucosal co-delivery of IL-1β, which significantly increased antibody and T cell responses in inbred Babraham pigs. Another group of outbred pigs was first exposed to pH1N1 as an alternative means of inducing heterosubtypic immunity and were subsequently challenged with H3N2. Although both prior infection and adenoviral vector immunization induced strong T-cell responses against the conserved NP protein, none of the treatment groups demonstrated increased protection against the heterologous H3N2 challenge. Ad-HA/NP+Ad-IL-1β immunization increased lung pathology, although viral load was unchanged. These data indicate that heterotypic immunity may be difficult to achieve in pigs and the immunological mechanisms may differ from those in small animal models. Caution should be applied in extrapolating from a single model to humans., Competing Interests: CT and SiS are employees of SIRION Biotech GmbH, a wholly-owned subsidiary of PerkinElmer Inc.. CT receives shares from the company. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Schmidt, Paudyal, Villanueva-Hernández, Mcnee, Vatzia, Carr, Schmidt, Mccarron, Martini, Schroedel, Thirion, Waters, Salguero, Gerner, Tenbusch and Tchilian.)
- Published
- 2023
- Full Text
- View/download PDF
37. Immunization with matrix-, nucleoprotein and neuraminidase protects against H3N2 influenza challenge in pH1N1 pre-exposed pigs.
- Author
-
Vatzia E, Feest K, McNee A, Manjegowda T, Carr BV, Paudyal B, Chrun T, Maze EA, Mccarron A, Morris S, Everett HE, MacLoughlin R, Salguero FJ, Lambe T, Gilbert SC, and Tchilian E
- Abstract
There is an urgent need for influenza vaccines providing broader protection that may decrease the need for annual immunization of the human population. We investigated the efficacy of heterologous prime boost immunization with chimpanzee adenovirus (ChAdOx2) and modified vaccinia Ankara (MVA) vectored vaccines, expressing conserved influenza virus nucleoprotein (NP), matrix protein 1 (M1) and neuraminidase (NA) in H1N1pdm09 pre-exposed pigs. We compared the efficacy of intra-nasal, aerosol and intra-muscular vaccine delivery against H3N2 influenza challenge. Aerosol prime boost immunization induced strong local lung T cell and antibody responses and abrogated viral shedding and lung pathology following H3N2 challenge. In contrast, intramuscular immunization induced powerful systemic responses and weak local lung responses but also abolished lung pathology and reduced viral shedding. These results provide valuable insights into the development of a broadly protective influenza vaccine in a highly relevant large animal model and will inform future vaccine and clinical trial design., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
38. Fc-Mediated Functions of Porcine IgG Subclasses.
- Author
-
Paudyal B, Mwangi W, Rijal P, Schwartz JC, Noble A, Shaw A, Sealy JE, Bonnet-Di Placido M, Graham SP, Townsend A, Hammond JA, and Tchilian E
- Subjects
- Animals, Antibodies, Monoclonal, Antigen-Antibody Complex, Complement System Proteins, Phagocytosis, Swine, Antibody-Dependent Cell Cytotoxicity, Immunoglobulin G
- Abstract
The pig is an important agricultural species and powerful biomedical model. We have established the pig, a large natural host animal for influenza with many physiological similarities to humans, as a robust model for testing the therapeutic potential of monoclonal antibodies. Antibodies provide protection through neutralization and recruitment of innate effector functions through the Fc domain. However very little is known about the Fc-mediated functions of porcine IgG subclasses. We have generated 8 subclasses of two porcine monoclonal anti influenza hemagglutinin antibodies. We characterized their ability to activate complement, trigger cytotoxicity and phagocytosis by immune cells and assayed their binding to monocytes, macrophages, and natural killer cells. We show that IgG1, IgG2a, IgG2b, IgG2c and IgG4 bind well to targeted cell types and mediate complement mediated cellular cytotoxicity (CDCC), antibody dependent cellular cytotoxicity (ADCC) and antibody mediated cell phagocytosis (ADCP). IgG5b and IgG5c exhibited weak binding and variable and poor functional activity. Immune complexes of porcine IgG3 did not show any Fc-mediated functions except for binding to monocytes and macrophages and weak binding to NK cells. Interestingly, functionally similar porcine IgG subclasses clustered together in the genome. These novel findings will enhance the utility of the pig model for investigation of therapeutic antibodies., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Paudyal, Mwangi, Rijal, Schwartz, Noble, Shaw, Sealy, Bonnet-Di Placido, Graham, Townsend, Hammond and Tchilian.)
- Published
- 2022
- Full Text
- View/download PDF
39. Porcine Respiratory Coronavirus as a Model for Acute Respiratory Coronavirus Disease.
- Author
-
Keep S, Carr BV, Lean FZX, Fones A, Newman J, Dowgier G, Freimanis G, Vatzia E, Polo N, Everest H, Webb I, Mcnee A, Paudyal B, Thakur N, Nunez A, MacLoughlin R, Maier H, Hammond J, Bailey D, Waters R, Charleston B, Tuthill T, Britton P, Bickerton E, and Tchilian E
- Subjects
- Animals, SARS-CoV-2, Swine, COVID-19, Porcine Respiratory Coronavirus, Swine Diseases, Transmissible gastroenteritis virus
- Abstract
In the light of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic, we have developed a porcine respiratory coronavirus (PRCV) model for in depth mechanistic evaluation of the pathogenesis, virology and immune responses of this important family of viruses. Pigs are a large animal with similar physiology and immunology to humans and are a natural host for PRCV. Four PRCV strains were investigated and shown to induce different degrees of lung pathology. Importantly, although all four strains replicated equally well in porcine cell lines in vitro and in the upper respiratory tract in vivo , PRCV strains causing more severe lung pathology were also able to replicate in ex vivo tracheal organ cultures as well as in vivo in the trachea and lung. The time course of infection of PRCV 135, which caused the most severe pulmonary pathology, was investigated. Virus was shed from the upper respiratory tract until day 10 post infection, with infection of the respiratory mucosa, as well as olfactory and sustentacular cells, providing an excellent model to study upper respiratory tract disease in addition to the commonly known lower respiratory tract disease from PRCV. Infected animals made antibody and T cell responses that cross reacted with the four PRCV strains and Transmissible Gastroenteritis Virus. The antibody response was reproduced in vitro in organ cultures. Comparison of mechanisms of infection and immune control in pigs infected with PRCVs of differing pathogenicity with human data from SARS-CoV-2 infection and from our in vitro organ cultures, will enable key events in coronavirus infection and disease pathogenesis to be identified., Competing Interests: RM is an employee of company Aerogen. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Keep, Carr, Lean, Fones, Newman, Dowgier, Freimanis, Vatzia, Polo, Everest, Webb, Mcnee, Paudyal, Thakur, Nunez, MacLoughlin, Maier, Hammond, Bailey, Waters, Charleston, Tuthill, Britton, Bickerton and Tchilian.)
- Published
- 2022
- Full Text
- View/download PDF
40. Spatial, temporal and molecular dynamics of swine influenza virus-specific CD8 tissue resident memory T cells.
- Author
-
Martini V, Edmans M, Gubbins S, Jayaraman S, Paudyal B, Morgan S, McNee A, Morin T, Rijal P, Gerner W, Sewell AK, Inoue R, Bailey M, Connelley T, Charleston B, Townsend A, Beverley P, and Tchilian E
- Subjects
- Animals, CD8-Positive T-Lymphocytes, Epitopes, Humans, Immunologic Memory, Memory T Cells, Molecular Dynamics Simulation, Swine, Influenza A virus, Influenza, Human, Orthomyxoviridae Infections
- Abstract
For the first time we have defined naïve, central memory, effector memory and differentiated effector porcine CD8 T cells and analyzed their distribution in lymphoid and respiratory tissues after influenza infection or immunization, using peptide-MHC tetramers of three influenza nucleoprotein (NP) epitopes. The hierarchy of response to the three epitopes changes during the response in different tissues. Most NP-specific CD8 T cells in broncho-alveolar lavage (BAL) and lung are tissue resident memory cells (TRM) that express CD69 and downregulate CD45RA and CCR7. NP-specific cells isolated from BAL express genes characteristic of TRM, but gene expression differs at 7, 21 and 63 days post infection. In all tissues the frequency of NP-specific CD8 cells declines over 63 days almost to background levels but is best maintained in BAL. The kinetic of influenza specific memory CD8 T cell in this natural host species differs from that in small animal models., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
41. Low Dose Pig Anti-Influenza Virus Monoclonal Antibodies Reduce Lung Pathology but Do Not Prevent Virus Shedding.
- Author
-
Paudyal B, McNee A, Rijal P, Carr BV, Nunez A, McCauley J, Daniels RS, Townsend AR, Hammond JA, and Tchilian E
- Subjects
- Administration, Intravenous, Animals, Bronchoalveolar Lavage Fluid virology, Disease Models, Animal, Female, Hemagglutinin Glycoproteins, Influenza Virus immunology, Humans, Influenza A Virus, H1N1 Subtype immunology, Influenza, Human immunology, Influenza, Human transmission, Influenza, Human virology, Lung drug effects, Lung immunology, Lung pathology, Lung virology, Nasal Mucosa drug effects, Nasal Mucosa pathology, Nasal Mucosa virology, Sus scrofa, Viral Load immunology, Virus Shedding drug effects, Virus Shedding immunology, Antibodies, Monoclonal administration & dosage, Antibodies, Viral administration & dosage, Influenza A Virus, H1N1 Subtype isolation & purification, Influenza, Human drug therapy
- Abstract
We have established the pig, a large natural host animal for influenza, with many physiological similarities to humans, as a robust model for testing the therapeutic potential of monoclonal antibodies (mAbs). In this study we demonstrated that prophylactic intravenous administration of 15 mg/kg of porcine mAb pb18, against the K160-163 site of the hemagglutinin, significantly reduced lung pathology and nasal virus shedding and eliminated virus from the lung of pigs following H1N1pdm09 challenge. When given at 1 mg/kg, pb18 significantly reduced lung pathology and lung and BAL virus loads, but not nasal shedding. Similarly, when pb18 was given in combination with pb27, which recognized the K130 site, at 1 mg/kg each, lung virus load and pathology were reduced, although without an apparent additive or synergistic effect. No evidence for mAb driven virus evolution was detected. These data indicate that intravenous administration of high doses was required to reduce nasal virus shedding, although this was inconsistent and seldom complete. In contrast, the effect on lung pathology and lung virus load is consistent and is also seen at a one log lower dose, strongly indicating that a lower dose might be sufficient to reduce severity of disease, but for prevention of transmission other measures would be needed., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Paudyal, McNee, Rijal, Carr, Nunez, McCauley, Daniels, Townsend, Hammond and Tchilian.)
- Published
- 2021
- Full Text
- View/download PDF
42. Simultaneous Infection With Porcine Reproductive and Respiratory Syndrome and Influenza Viruses Abrogates Clinical Protection Induced by Live Attenuated Porcine Reproductive and Respiratory Syndrome Vaccination.
- Author
-
Chrun T, Maze EA, Vatzia E, Martini V, Paudyal B, Edmans MD, McNee A, Manjegowda T, Salguero FJ, Wanasen N, Koonpaew S, Graham SP, and Tchilian E
- Subjects
- Animals, Antibodies, Viral biosynthesis, Coinfection immunology, Coinfection virology, Cytokines biosynthesis, Cytokines genetics, Datasets as Topic, Dogs, Female, Madin Darby Canine Kidney Cells, Orthomyxoviridae Infections complications, Orthomyxoviridae Infections virology, Porcine Reproductive and Respiratory Syndrome virology, Swine, Vaccination veterinary, Vaccine Efficacy, Vaccines, Attenuated immunology, Viral Load, Viremia prevention & control, Viremia virology, Coinfection veterinary, Influenza A Virus, H3N2 Subtype immunology, Orthomyxoviridae Infections immunology, Porcine Reproductive and Respiratory Syndrome prevention & control, Porcine respiratory and reproductive syndrome virus immunology, Viral Vaccines immunology
- Abstract
The porcine respiratory disease complex (PRDC) is responsible for significant economic losses in the pig industry worldwide. Porcine reproductive and respiratory syndrome virus (PRRSV) and swine influenza virus are major viral contributors to PRDC. Vaccines are cost-effective measures for controlling PRRS, however, their efficacy in the context of co-infections has been poorly investigated. In this study, we aimed to determine the effect of PRRSV-2 and swine influenza H3N2 virus co-infection on the efficacy of PRRSV modified live virus (MLV) vaccination, which is widely used in the field. Following simultaneous challenge with contemporary PRRSV-2 and H3N2 field isolates, we found that the protective effect of PRRS MLV vaccination on clinical disease and pathology was abrogated, although viral load was unaffected and antibody responses were enhanced. In contrast, co-infection in non-immunized animals reduced PRRSV-2 viremia and H3N2 virus load in the upper respiratory tract and potentiated T cell responses against both PRRSV-2 and H3N2 in the lung. Further analysis suggested that an upregulation of inhibitory cytokines gene expression in the lungs of vaccinated pigs may have influenced responses to H3N2 and PRRSV-2. These findings provide important insights into the effect of viral co-infections on PRRS vaccine efficacy that may help identify more effective vaccination strategies against PRDC in the field., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Chrun, Maze, Vatzia, Martini, Paudyal, Edmans, McNee, Manjegowda, Salguero, Wanasen, Koonpaew, Graham and Tchilian.)
- Published
- 2021
- Full Text
- View/download PDF
43. Respiratory and Intramuscular Immunization With ChAdOx2-NPM1-NA Induces Distinct Immune Responses in H1N1pdm09 Pre-Exposed Pigs.
- Author
-
Vatzia E, Allen ER, Manjegowda T, Morris S, McNee A, Martini V, Kaliath R, Ulaszewska M, Boyd A, Paudyal B, Carr VB, Chrun T, Maze E, MacLoughlin R, van Diemen PM, Everett HE, Lambe T, Gilbert SC, and Tchilian E
- Subjects
- Adenoviridae genetics, Aerosols, Animals, Cytokines biosynthesis, Influenza Vaccines administration & dosage, Neuraminidase immunology, Nucleocapsid Proteins immunology, Swine, Vaccination, Virus Shedding, Antibodies, Viral blood, Influenza A Virus, H1N1 Subtype immunology, Influenza Vaccines immunology, Viral Matrix Proteins immunology
- Abstract
There is a critical need to develop superior influenza vaccines that provide broader protection. Influenza vaccines are traditionally tested in naive animals, although humans are exposed to influenza in the first years of their lives, but the impact of prior influenza exposure on vaccine immune responses has not been well studied. Pigs are an important natural host for influenza, are a source of pandemic viruses, and are an excellent model for human influenza. Here, we investigated the immunogenicity of the ChAdOx2 viral vectored vaccine, expressing influenza nucleoprotein, matrix protein 1, and neuraminidase in H1N1pdm09 pre-exposed pigs. We evaluated the importance of the route of administration by comparing intranasal, aerosol, and intramuscular immunizations. Aerosol delivery boosted the local lung T-cell and antibody responses, while intramuscular immunization boosted peripheral blood immunity. These results will inform how best to deliver vaccines in order to harness optimal protective immunity., Competing Interests: SG is co-founder of Vaccitech and named as an inventor on a patent covering use of ChAdOx2-vectored vaccines. TL is a consultant to Vaccitech. RM is an employee of Aerogen Limited and is a named inventor on inhaled vaccine delivery system patents. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Vatzia, Allen, Manjegowda, Morris, McNee, Martini, Kaliath, Ulaszewska, Boyd, Paudyal, Carr, Chrun, Maze, MacLoughlin, van Diemen, Everett, Lambe, Gilbert and Tchilian.)
- Published
- 2021
- Full Text
- View/download PDF
44. Changes in the Nasal Microbiota of Pigs Following Single or Co-Infection with Porcine Reproductive and Respiratory Syndrome and Swine Influenza A Viruses.
- Author
-
Chrun T, Leng J, La Ragione RM, Graham SP, and Tchilian E
- Abstract
Host-microbiota interactions are important in shaping immune responses that have the potential to influence the outcome of pathogen infection. However, most studies have focused on the gut microbiota and its possible association with disease outcome, while the role of the nasal microbiota and respiratory pathogen infection has been less well studied. Here we examined changes in the composition of the nasal microbiota of pigs following experimental infection with porcine reproductive and respiratory syndrome virus 2 (PRRSV-2), swine influenza A H3N2 virus (H3N2) or both viruses. DNA extracted from nasal swabs were subjected to 16S rRNA sequencing to study the composition of the nasal microbiota. Bacterial richness fluctuated in all groups, with a slight reduction in pigs singly infected with PRRSV-2 and H3N2 during the first 5 days of infection compared to uninfected controls. In contrast, nasal bacterial richness remained relatively stable after PRRSV-2/H3N2 co-infection. PRRSV-2 and H3N2, alone or in combination differentially altered the abundance and distribution of bacterial families. Single and co-infection with PRRSV-2 or H3N2 was associated with the expansion of the Neisseriaceae family. A positive correlation between H3N2 viral load and the relative abundance of the Neisseriaceae was observed. However, further mechanistic studies are required to understand the significance of the changes in specific bacterial families following these viral infections.
- Published
- 2021
- Full Text
- View/download PDF
45. Correction: Protective porcine influenza virus-specific monoclonal antibodies recognize similar haemagglutinin epitopes as humans.
- Author
-
Holzer B, Rijal P, McNee A, Paudyal B, Martini V, Clark B, Manjegowda T, Salguero FJ, Bessell E, Schwartz JC, Moffat K, Pedrera M, Graham SP, Noble A, Placido MB, La Ragione RM, Mwangi W, Beverley P, McCauley JW, Daniels RS, Hammond JA, Townsend AR, and Tchilian E
- Abstract
[This corrects the article DOI: 10.1371/journal.ppat.1009330.].
- Published
- 2021
- Full Text
- View/download PDF
46. Correction: Simultaneous Aerosol and Intramuscular Immunization with Influenza Vaccine Induces Powerful Protective Local T Cell and Systemic Antibody Immune Responses in Pigs.
- Author
-
Martini V, Paudyal B, Chrun T, McNee A, Edmans M, Atangana Maze E, Clark B, Nunez A, Dolton G, Sewell A, Beverley P, MacLoughlin R, Townsend A, and Tchilian E
- Published
- 2021
- Full Text
- View/download PDF
47. Identification and Phenotype of MAIT Cells in Cattle and Their Response to Bacterial Infections.
- Author
-
Edmans MD, Connelley TK, Jayaraman S, Vrettou C, Vordermeier M, Mak JYW, Liu L, Fairlie DP, Maze EA, Chrun T, Klenerman P, Eckle SBG, Tchilian E, and Benedictus L
- Subjects
- Animals, Cytokines pharmacology, Female, Histocompatibility Antigens Class I immunology, Humans, Male, Mice, Minor Histocompatibility Antigens immunology, Phenotype, Ribitol analogs & derivatives, Ribitol pharmacology, Uracil analogs & derivatives, Uracil pharmacology, Bacterial Infections immunology, Cattle immunology, Mucosal-Associated Invariant T Cells immunology
- Abstract
Mucosal-associated invariant T (MAIT) cells are a population of innate-like T cells that utilize a semi-invariant T cell receptor (TCR) α chain and are restricted by the highly conserved antigen presenting molecule MR1. MR1 presents microbial riboflavin biosynthesis derived metabolites produced by bacteria and fungi. Consistent with their ability to sense ligands derived from bacterial sources, MAIT cells have been associated with the immune response to a variety of bacterial infections, such as Mycobacterium spp., Salmonella spp. and Escherichia coli . To date, MAIT cells have been studied in humans, non-human primates and mice. However, they have only been putatively identified in cattle by PCR based methods; no phenotypic or functional analyses have been performed. Here, we identified a MAIT cell population in cattle utilizing MR1 tetramers and high-throughput TCR sequencing. Phenotypic analysis of cattle MAIT cells revealed features highly analogous to those of MAIT cells in humans and mice, including expression of an orthologous TRAV1-TRAJ33 TCR α chain, an effector memory phenotype irrespective of tissue localization, and expression of the transcription factors PLZF and EOMES. We determined the frequency of MAIT cells in peripheral blood and multiple tissues, finding that cattle MAIT cells are enriched in mucosal tissues as well as in the mesenteric lymph node. Cattle MAIT cells were responsive to stimulation by 5-OP-RU and riboflavin biosynthesis competent bacteria in vitro . Furthermore, MAIT cells in milk increased in frequency in cows with mastitis. Following challenge with virulent Mycobacterium bovis , a causative agent of bovine tuberculosis and a zoonosis, peripheral blood MAIT cells expressed higher levels of perforin. Thus, MAIT cells are implicated in the immune response to two major bacterial infections in cattle. These data suggest that MAIT cells are functionally highly conserved and that cattle are an excellent large animal model to study the role of MAIT cells in important zoonotic infections., Competing Interests: SE, JM, LL, and DF are inventors on patents describing MR1 tetramers and MR1–ligand complexes. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Edmans, Connelley, Jayaraman, Vrettou, Vordermeier, Mak, Liu, Fairlie, Maze, Chrun, Klenerman, Eckle, Tchilian and Benedictus.)
- Published
- 2021
- Full Text
- View/download PDF
48. Protective porcine influenza virus-specific monoclonal antibodies recognize similar haemagglutinin epitopes as humans.
- Author
-
Holzer B, Rijal P, McNee A, Paudyal B, Martini V, Clark B, Manjegowda T, Salguero FJ, Bessell E, Schwartz JC, Moffat K, Pedrera M, Graham SP, Noble A, Bonnet-Di Placido M, La Ragione RM, Mwangi W, Beverley P, McCauley JW, Daniels RS, Hammond JA, Townsend AR, and Tchilian E
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antibodies, Viral immunology, Hemagglutinins immunology, Hemagglutinins pharmacology, Humans, Influenza A Virus, H1N1 Subtype drug effects, Influenza A Virus, H1N1 Subtype immunology, Influenza A virus drug effects, Influenza A virus immunology, Influenza Vaccines immunology, Influenza, Human virology, Swine, Antibodies, Viral pharmacology, Epitopes immunology, Hemagglutinin Glycoproteins, Influenza Virus immunology, Influenza, Human drug therapy
- Abstract
Pigs are natural hosts for the same subtypes of influenza A viruses as humans and integrally involved in virus evolution with frequent interspecies transmissions in both directions. The emergence of the 2009 pandemic H1N1 virus illustrates the importance of pigs in evolution of zoonotic strains. Here we generated pig influenza-specific monoclonal antibodies (mAbs) from H1N1pdm09 infected pigs. The mAbs recognized the same two major immunodominant haemagglutinin (HA) epitopes targeted by humans, one of which is not recognized by post-infection ferret antisera that are commonly used to monitor virus evolution. Neutralizing activity of the pig mAbs was comparable to that of potent human anti-HA mAbs. Further, prophylactic administration of a selected porcine mAb to pigs abolished lung viral load and greatly reduced lung pathology but did not eliminate nasal shedding of virus after H1N1pdm09 challenge. Hence mAbs from pigs, which target HA can significantly reduce disease severity. These results, together with the comparable sizes of pigs and humans, indicate that the pig is a valuable model for understanding how best to apply mAbs as therapy in humans and for monitoring antigenic drift of influenza viruses in humans, thereby providing information highly relevant to making influenza vaccine recommendations., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
49. Magnitude and Kinetics of T Cell and Antibody Responses During H1N1pdm09 Infection in Inbred Babraham Pigs and Outbred Pigs.
- Author
-
Edmans M, McNee A, Porter E, Vatzia E, Paudyal B, Martini V, Gubbins S, Francis O, Harley R, Thomas A, Burt R, Morgan S, Fuller A, Sewell A, Charleston B, Bailey M, and Tchilian E
- Subjects
- Animals, B-Lymphocytes immunology, B-Lymphocytes metabolism, B-Lymphocytes virology, Cytokines metabolism, Disease Models, Animal, Histocompatibility Antigens Class I genetics, Histocompatibility Antigens Class I immunology, Host-Pathogen Interactions, Inbreeding, Influenza A Virus, H1N1 Subtype pathogenicity, Kinetics, Orthomyxoviridae Infections blood, Orthomyxoviridae Infections genetics, Orthomyxoviridae Infections immunology, Receptors, Antigen, T-Cell, alpha-beta metabolism, Receptors, Antigen, T-Cell, gamma-delta metabolism, Species Specificity, Sus scrofa, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets metabolism, Viral Load, Virus Shedding, Antibodies, Viral blood, Influenza A Virus, H1N1 Subtype immunology, Orthomyxoviridae Infections virology, T-Lymphocyte Subsets virology
- Abstract
We have used the pig, a large natural host animal for influenza with many physiological similarities to humans, to characterize αβ, γδ T cell and antibody (Ab) immune responses to the 2009 pandemic H1N1 virus infection. We evaluated the kinetic of virus infection and associated response in inbred Babraham pigs with identical MHC (Swine Leucocyte Antigen) and compared them to commercial outbred animals. High level of nasal virus shedding continued up to days 4 to 5 post infection followed by a steep decline and clearance of virus by day 9. Adaptive T cell and Ab responses were detectable from days 5 to 6 post infection reaching a peak at 9 to 14 days. γδ T cells produced cytokines ex vivo at day 2 post infection, while virus reactive IFNγ producing γδ T cells were detected from day 7 post infection. Analysis of NP tetramer specific and virus specific CD8 and CD4 T cells in blood, lung, lung draining lymph nodes, and broncho-alveolar lavage (BAL) showed clear differences in cytokine production between these tissues. BAL contained the most highly activated CD8, CD4, and γδ T cells producing large amounts of cytokines, which likely contribute to elimination of virus. The weak response in blood did not reflect the powerful local lung immune responses. The immune response in the Babraham pig following H1N1pdm09 influenza infection was comparable to that of outbred animals. The ability to utilize these two swine models together will provide unparalleled power to analyze immune responses to influenza., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Edmans, McNee, Porter, Vatzia, Paudyal, Martini, Gubbins, Francis, Harley, Thomas, Burt, Morgan, Fuller, Sewell, Charleston, Bailey and Tchilian.)
- Published
- 2021
- Full Text
- View/download PDF
50. Simultaneous Aerosol and Intramuscular Immunization with Influenza Vaccine Induces Powerful Protective Local T Cell and Systemic Antibody Immune Responses in Pigs.
- Author
-
Martini V, Paudyal B, Chrun T, McNee A, Edmans M, Atangana Maze E, Clark B, Nunez A, Dolton G, Sewell A, Beverley P, MacLoughlin R, Townsend A, and Tchilian E
- Subjects
- Aerosols, Animals, Antibody Formation, Disease Models, Animal, Disease Resistance, Humans, Immunity, Cellular, Immunization, Injections, Intramuscular, Swine, Influenza A Virus, H1N1 Subtype physiology, Influenza Vaccines immunology, Influenza, Human immunology, Orthomyxoviridae Infections immunology, T-Lymphocytes immunology
- Abstract
A vaccine providing both powerful Ab and cross-reactive T cell immune responses against influenza viruses would be beneficial for both humans and pigs. In this study, we evaluated i.m., aerosol (Aer), and simultaneous systemic and respiratory immunization (SIM) by both routes in Babraham pigs, using the single cycle candidate influenza vaccine S-FLU. After prime and boost immunization, pigs were challenged with H1N1pdm09 virus. i.m.-immunized pigs generated a high titer of neutralizing Abs but poor T cell responses, whereas Aer induced powerful respiratory tract T cell responses but a low titer of Abs. SIM pigs combined high Ab titers and strong local T cell responses. SIM showed the most complete suppression of virus shedding and the greatest improvement in pathology. We conclude that SIM regimes for immunization against respiratory pathogens warrant further study., (Copyright © 2021 The Authors.)
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.