1
|
Do KTH, Willenzon S, Ristenpart J, Janssen A, Volz A, Sutter G, Förster R, Bošnjak B. The effect of Toll-like receptor agonists on the immunogenicity of MVA-SARS-2-S vaccine after intranasal administration in mice. Front Cell Infect Microbiol 2023; 13:1259822. [PMID: 37854858 PMCID: PMC10580083 DOI: 10.3389/fcimb.2023.1259822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 09/14/2023] [Indexed: 10/20/2023] Open
Abstract
Background and aims Modified Vaccinia virus Ankara (MVA) represents a promising vaccine vector for respiratory administration to induce protective lung immunity including tertiary lymphoid structure, the bronchus-associated lymphoid tissue (BALT). However, MVA expressing the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Spike protein (MVA-SARS-2-S) required prime-boost administration to induce high titers of anti-Spike antibodies in serum and bronchoalveolar lavage (BAL). As the addition of adjuvants enables efficient tailoring of the immune responses even to live vaccines, we tested whether Toll-like receptor (TLR)-agonists affect immune responses induced by a single dose of intranasally applied MVA-SARS-2-S. Methods We intranasally immunized C57BL/6 mice with MVA-SARS-2-S vaccine in the presence of either TLR3 agonist polyinosinic polycytidylic acid [poly(I:C)], TLR4 agonist bacterial lipopolysaccharide (LPS) from Escherichia coli, or TLR9 agonist CpG oligodeoxynucleotide (CpG ODN) 1826. At different time-points after immunization, we analyzed induced immune responses using flow cytometry, immunofluorescent microscopy, and ELISA. Results TLR agonists had profound effects on MVA-SARS-2-S-induced immune responses. At day 1 post intranasal application, the TLR4 agonist significantly affected MVA-induced activation of dendritic cells (DCs) within the draining bronchial lymph nodes, increasing the ratio of CD11b+CD86+ to CD103+CD86+ DCs. Nevertheless, the number of Spike-specific CD8+ T cells within the lungs at day 12 after vaccination was increased in mice that received MVA-SARS-2-S co-administered with TLR3 but not TLR4 agonists. TLR9 agonist did neither significantly affect MVA-induced DC activation nor the induction of Spike-specific CD8+ T cells but reduced both number and size of bronchus-associated lymphoid tissue. Surprisingly, the addition of all TLR agonists failed to boost the levels of Spike-specific antibodies in serum and bronchoalveolar lavage. Conclusions Our study indicates a potential role of TLR-agonists as a tool to modulate immune responses to live vector vaccines. Particularly TLR3 agonists hold a promise to potentiate MVA-induced cellular immune responses. On the other hand, additional research is necessary to identify optimal combinations of agonists that could enhance MVA-induced humoral responses.
Collapse
Affiliation(s)
- Kim Thi Hoang Do
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | | | | | - Anika Janssen
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Asisa Volz
- Institute for Virology, University of Veterinary Medicine Hannover, Hannover, Germany
- German Centre for Infection Research (DZIF), Munich, Germany
| | - Gerd Sutter
- German Centre for Infection Research (DZIF), Munich, Germany
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximiliam University (LMU) Munich, Munich, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- German Centre for Infection Research (DZIF), Hannover, Germany
| | - Berislav Bošnjak
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
2
|
Yersinia pestis Antigen F1 but Not LcrV Induced Humoral and Cellular Immune Responses in Humans Immunized with Live Plague Vaccine-Comparison of Immunoinformatic and Immunological Approaches. Vaccines (Basel) 2020; 8:vaccines8040698. [PMID: 33228200 PMCID: PMC7712656 DOI: 10.3390/vaccines8040698] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/01/2020] [Accepted: 11/16/2020] [Indexed: 12/21/2022] Open
Abstract
The recent progress in immunoinformatics provided the basis for an accelerated development of target-specific peptide vaccines as an alternative to the traditional vaccine concept. However, there is still limited information on whether the in silico predicted immunoreactive epitopes correspond to those obtained from the actual experiments. Here, humoral and cellular immune responses to two major Yersinia pestis protective antigens, F1 and LcrV, were studied in human donors immunized with the live plague vaccine (LPV) based on the attenuated Y. pestis strain EV line NIIEG. The F1 antigen provided modest specific cellular (mixed T helper 1 (Th1)/Th2 type) and humoral immune responses in vaccinees irrespective of the amount of annual vaccinations and duration of the post-vaccination period. The probing of the F1 overlapping peptide library with the F1-positive sera revealed the presence of seven linear B cell epitopes, which were all also predicted by in silico assay. The immunoinformatics study evaluated their antigenicity, toxicity, and allergenic properties. The epitope TSQDGNNH was mostly recognized by the sera from recently vaccinated donors rather than antibodies from those immunized decades ago, suggesting the usefulness of this peptide for differentiation between recent and long-term vaccinations. The in silico analysis predicted nine linear LcrV-specific B-cell epitopes; however, weak antibody and cellular immune responses prevented their experimental evaluation, indicating that LcrV is a poor marker of successful vaccination. No specific Th17 immune response to either F1 or LcrV was detected, and there were no detectable serum levels of F1-specific immunoglobulin A (IgA) in vaccinees. Overall, the general approach validated in the LPV model could be valuable for the rational design of vaccines against other neglected and novel emerging infections with high pandemic potency.
Collapse
|
3
|
Hamaoka S, Naito Y, Katoh H, Shimizu M, Kinoshita M, Akiyama K, Kainuma A, Moriyama K, Ishii KJ, Sawa T. Efficacy comparison of adjuvants in PcrV vaccine against Pseudomonas aeruginosa pneumonia. Microbiol Immunol 2017; 61:64-74. [PMID: 28370521 DOI: 10.1111/1348-0421.12467] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/16/2017] [Accepted: 01/19/2017] [Indexed: 11/27/2022]
Abstract
Vaccination against the type III secretion system of P. aeruginosa is a potential prophylactic strategy for reducing the incidence and improving the poor prognosis of P. aeruginosa pneumonia. In this study, the efficacies of three different adjuvants, Freund's adjuvant (FA), aluminum hydroxide (alum) and CpG oligodeoxynucleotide (ODN), were examined from the viewpoint of inducing PcrV-specific immunity against virulent P. aeruginosa. Mice that had been immunized intraperitoneally with recombinant PcrV formulated with one of the above adjuvants were challenged intratracheally with a lethal dose of P. aeruginosa. The PcrV-FA immunized group attained a survival rate of 91%, whereas the survival rates of the PcrV-alum and PcrV-CpG groups were 73% and 64%, respectively. In terms of hypothermia recovery after bacterial instillation, PcrV-alum was the most protective, followed by PcrV-FA and PcrV-CpG. The lung edema index was lower in the PcrV-CpG vaccination group than in the other groups. PcrV-alum immunization was associated with the greatest decrease in myeloperoxidase in infected lungs, and also decreased the number of lung bacteria to a similar number as in the PcrV-FA group. There was less neutrophil recruitment in the lungs of mice vaccinated with PcrV-alum or PcrV-CpG than in those of mice vaccinated with PcrV-FA or PcrV alone. Overall, in terms of mouse survival the PcrV-CpG vaccine, which could be a relatively safe next-generation vaccine, showed a comparable effect to the PcrV-alum vaccine.
Collapse
Affiliation(s)
- Saeko Hamaoka
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yoshifumi Naito
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Hideya Katoh
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Masaru Shimizu
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Mao Kinoshita
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Koichi Akiyama
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Atsushi Kainuma
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Kiyoshi Moriyama
- Department of Anesthesiology, School of Medicine, Kyorin University, Mitaka 181-8611, Japan
| | - Ken J Ishii
- Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan.,Laboratory of Vaccine Science, Immunology Frontier Research Center, World Premier International Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| | - Teiji Sawa
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
4
|
Ferreirinha P, Correia A, Teixeira-Coelho M, Osório H, Teixeira L, Rocha A, Vilanova M. Mucosal immunization confers long-term protection against intragastrically established Neospora caninum infection. Vaccine 2016; 34:6250-6258. [PMID: 27814932 DOI: 10.1016/j.vaccine.2016.10.056] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/07/2016] [Accepted: 10/24/2016] [Indexed: 11/25/2022]
Abstract
Neospora caninum is an obligate intracellular protozoan parasite responsible for heavy economic losses in dairy and beef cattle farms worldwide. Although vaccination is widely regarded as the preferable strategy to prevent neosporosis no commercial vaccine is currently available. We have previously shown that intranasal immunization with an N. caninum antigen extract enriched in hydrophobic proteins plus CpG adjuvant protected mice against intragastrically established neosporosis. Nevertheless, the antigen specificity as well as the long-term protective effect of this immunization strategy were not determined. Here, we show that the protective effect of this intranasal immunization procedure lasted for at least 20weeks. Protection was accompanied by long-lasting elevated levels of parasite-specific serum IgG and intestinal IgA. Moreover, spleen and mesenteric lymph node cells obtained from non-infected long-term immunized mice responded by producing interferon-γ following in vitro parasite-antigen recall. Analysis of serum IgG and intestinal IgA antibody reactivity in immunized mice identified dense granule antigen 7 (NcGRA7) and microneme associated protein 1 (NcMIC1) as immunodominant antigens respectively recognized by those antibody fractions. In summary, this work shows that a previously reported mucosal immunization strategy against N. caninum infection established through the gastrointestinal tract is effective in the long term.
Collapse
Affiliation(s)
- Pedro Ferreirinha
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Alexandra Correia
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Maria Teixeira-Coelho
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Hugo Osório
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; IPATIMUP - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal; FMUP - Faculdade de Medicina da Universidade do Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Luzia Teixeira
- ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira n. 228, 4050-313 Porto, Portugal; UMIB - Unidade Multidisciplinar de Investigação Biomédica, Universidade do Porto, Portugal
| | - António Rocha
- ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira n. 228, 4050-313 Porto, Portugal
| | - Manuel Vilanova
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira n. 228, 4050-313 Porto, Portugal.
| |
Collapse
|
5
|
Singh AK, Kingston JJ, Gupta SK, Batra HV. Recombinant Bivalent Fusion Protein rVE Induces CD4+ and CD8+ T-Cell Mediated Memory Immune Response for Protection Against Yersinia enterocolitica Infection. Front Microbiol 2015; 6:1407. [PMID: 26733956 PMCID: PMC4679870 DOI: 10.3389/fmicb.2015.01407] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/25/2015] [Indexed: 11/16/2022] Open
Abstract
Studies investigating the correlates of immune protection against Yersinia infection have established that both humoral and cell mediated immune responses are required for the comprehensive protection. In our previous study, we established that the bivalent fusion protein (rVE) comprising immunologically active regions of Y. pestis LcrV (100–270 aa) and YopE (50–213 aa) proteins conferred complete passive and active protection against lethal Y. enterocolitica 8081 challenge. In the present study, cohort of BALB/c mice immunized with rVE or its component proteins rV, rE were assessed for cell mediated immune responses and memory immune protection against Y. enterocolitica 8081. rVE immunization resulted in extensive proliferation of both CD4 and CD8 T cell subsets; significantly high antibody titer with balanced IgG1: IgG2a/IgG2b isotypes (1:1 ratio) and up-regulation of both Th1 (TNF-α, IFN-γ, IL-2, and IL-12) and Th2 (IL-4) cytokines. On the other hand, rV immunization resulted in Th2 biased IgG response (11:1 ratio) and proliferation of CD4+ T-cell; rE group of mice exhibited considerably lower serum antibody titer with predominant Th1 response (1:3 ratio) and CD8+ T-cell proliferation. Comprehensive protection with superior survival (100%) was observed among rVE immunized mice when compared to the significantly lower survival rates among rE (37.5%) and rV (25%) groups when IP challenged with Y. enterocolitica 8081 after 120 days of immunization. Findings in this and our earlier studies define the bivalent fusion protein rVE as a potent candidate vaccine molecule with the capability to concurrently stimulate humoral and cell mediated immune responses and a proof of concept for developing efficient subunit vaccines against Gram negative facultative intracellular bacterial pathogens.
Collapse
Affiliation(s)
- Amit K Singh
- Department of Microbiology, Defence Food Research Laboratory, Defence Research and Development Organisation Mysore, India
| | - Joseph J Kingston
- Department of Microbiology, Defence Food Research Laboratory, Defence Research and Development Organisation Mysore, India
| | - Shishir K Gupta
- Department of Bioinformatics, Biocenter, University of Würzburg Würzburg, Germany
| | - Harsh V Batra
- Department of Microbiology, Defence Food Research Laboratory, Defence Research and Development Organisation Mysore, India
| |
Collapse
|
6
|
A Multiple Antigenic Peptide Mimicking Peptidoglycan Induced T Cell Responses to Protect Mice from Systemic Infection with Staphylococcus aureus. PLoS One 2015; 10:e0136888. [PMID: 26317210 PMCID: PMC4552945 DOI: 10.1371/journal.pone.0136888] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 08/10/2015] [Indexed: 12/11/2022] Open
Abstract
Due to the enormous capacity of Staphylococcus aureus to acquire antibiotic resistance, it becomes imperative to develop vaccines for decreasing the risk of its life-threatening infections. Peptidoglycan (PGN) is a conserved and major component of S. aureus cell wall. However, it has not been used as a vaccine candidate since it is a thymus-independent antigen. In this study, we synthesized a multiple antigenic peptide, named MAP27, which comprised four copies of a peptide that mimics the epitope of PGN. After immunization with MAP27 five times and boosting with heat-inactivated bacterium one time, anti-MAP27 serum bound directly to S. aureus or PGN. Immunization with MAP27 decreased the bacterial burden in organs of BALB/c mice and significantly prolonged their survival time after S. aureus lethal-challenge. The percentage of IFN-γ+CD3+ T cells and IL-17+CD4+ T cells in spleen, as well as the levels of IFN-γ, IL-17A/F and CCL3 in spleen and lung, significantly increased in the MAP27-immunized mice after infection. Moreover, in vitro incubation of heat-inactivated S. aureus with splenocytes isolated from MAP27-immunized mice stimulated the production of IFN-γ and IL-17A/F. Our findings demonstrated that MAP27, as a thymus-dependent antigen, is efficient at eliciting T cell-mediated responses to protect mice from S. aureus infection. This study sheds light on a possible strategy to design vaccines against S. aureus.
Collapse
|
7
|
Rai R, Das B, Choudhary N, Talukdar A, Rao DN. MAP of F1 and V antigens from Yersinia pestis astride innate and adaptive immune response. Microb Pathog 2015; 87:13-20. [PMID: 26188288 DOI: 10.1016/j.micpath.2015.07.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 07/05/2015] [Accepted: 07/13/2015] [Indexed: 01/31/2023]
Abstract
Yersinia pestis, a causative agent of plague, has a plethora of armors to fight against major components of innate immunity and survive within host cells. Dendritic cells and macrophages are important antigen presenting cells for effective immune response. This report is focused on the changes in DC activation and TLR2 and TLR4 expression on macrophages induced by MAP of F1 and V antigens of Y. pestis. F1 and V MAPs bear potential synthetic T and B cell epitopes from F1 and V protein respectively. We evaluated these parameters in DC's isolated from spleen and lamina propria and macrophages isolated from peritoneal lavage of mice after intranasal immunization. F1 MAP and V MAP significantly increased the expression of CD80 and CD86 on CD11c(+) dendritic cells isolated from spleen and lamina propria as well as intracellular IL-12 levels. Similarly, in macrophages derived from peritoneal cavity, the above formulation enhanced TLR2 and TLR4 expression. Again after in vitro stimulation with F1 and V MAP these macrophages produced significantly high IL12 and TNFα. The study clearly indicates involvement of DC and macrophages for efficient antigen presentation to immune cells. From this study we conclude that F1MAP and VMAP ameliorate innate immune mechanism. These two synthetic constructs exert their effect via TLR2 and TLR4, leading to the production of proinflammatory cytokines by macrophages and are able to increase DC activation, that could be helpful in generation of adaptive immunity as well as is important strong immune response.
Collapse
Affiliation(s)
- Reeta Rai
- Dept of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Baijnath Das
- Dept of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Nageshwar Choudhary
- Dept of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Ayantika Talukdar
- Dept of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | | |
Collapse
|
8
|
Gursel M, Klinman DM. Use of CpG Oligonucleotides as Mucosal Adjuvants. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00062-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
9
|
Pasiarski M, Rolinski J, Grywalska E, Stelmach-Goldys A, Korona-Glowniak I, Gozdz S, Hus I, Malm A. Antibody and plasmablast response to 13-valent pneumococcal conjugate vaccine in chronic lymphocytic leukemia patients--preliminary report. PLoS One 2014; 9:e114966. [PMID: 25506837 PMCID: PMC4266633 DOI: 10.1371/journal.pone.0114966] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 11/17/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Chronic lymphocytic leukemia (CLL) leads to significant immune system dysfunction. The predominant clinical presentation in 50% of patients involves recurrent, often severe, infections. Infections are also the most common (60-80%) cause of deaths in CLL patients. The scope of infections varies with the clinical stage of the disease. Treatment-naive patients typically present with respiratory tract infections caused by encapsulated bacteria Streptococcus pneumoniae and Haemophilus influenzae. Since 2012, the 13-valent pneumococcal conjugate vaccine (PCV13) has been recommended in the United States and some EU countries for pneumococcal infection prevention in patients with CLL (besides the long-standing standard, 23-valent pneumococcal polysaccharide vaccine, PPV23). The aim of this study was to compare the immune response to PCV13 in 24 previously untreated CLL patients and healthy subjects. METHODS Both groups were evaluated for: the levels of specific pneumococcal antibodies, the levels of IgG and IgG subclasses and selected peripheral blood lymphocyte subpopulations including the frequency of plasmablasts before and after immunization. RESULTS Adequate response to vaccination, defined as an at least two-fold increase in specific pneumococcal antibody titers versus pre-vaccination baseline titers, was found in 58.3% of CLL patients and 100% of healthy subjects. Both the CLL group and the control group demonstrated a statistically significant increase in the IgG2 subclass levels following vaccination (P = 0.0301). After vaccination, the frequency of plasmablasts was significantly lower (P<0.0001) in CLL patients in comparison to that in controls. Patients who responded to vaccination had lower clinical stage of CLL as well as higher total IgG, and IgG2 subclass levels. No significant vaccine-related side effects were observed. CONCLUSIONS PCV13 vaccination in CLL patients is safe and induces an effective immune response in a considerable proportion of patients. To achieve an optimal vaccination response, the administration of PCV13 is recommended as soon as possible following CLL diagnosis.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Antibodies, Bacterial/blood
- Antibodies, Bacterial/immunology
- B-Lymphocytes/immunology
- B-Lymphocytes/pathology
- Female
- Humans
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/blood
- Leukemia, Lymphocytic, Chronic, B-Cell/complications
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Male
- Middle Aged
- Pneumococcal Infections/blood
- Pneumococcal Infections/complications
- Pneumococcal Infections/immunology
- Pneumococcal Infections/prevention & control
- Pneumococcal Vaccines/immunology
- Pneumococcal Vaccines/therapeutic use
- Streptococcus pneumoniae/immunology
- Vaccines, Conjugate/immunology
- Vaccines, Conjugate/therapeutic use
Collapse
Affiliation(s)
- Marcin Pasiarski
- Department of Hematology, Holycross Cancer Center, Kielce, Poland
| | - Jacek Rolinski
- Department of Clinical Immunology and Immunotherapy, Medical University of Lublin, Lublin, Poland
- St. John’s Cancer Center, Lublin, Poland
| | - Ewelina Grywalska
- Department of Clinical Immunology and Immunotherapy, Medical University of Lublin, Lublin, Poland
- St. John’s Cancer Center, Lublin, Poland
| | | | | | - Stanislaw Gozdz
- Department of Chemotherapy and Clinical Oncology, Holycross Cancer Center, Kielce, Poland
- Faculty of Health Sciences, Jan Kochanowski University, Kielce, Poland
| | - Iwona Hus
- Department of Clinical Transplantology, Medical University of Lublin, Lublin, Poland
| | - Anna Malm
- Department of Pharmaceutical Microbiology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
10
|
Poojary NS, Ramlal S, Urs RM, Sripathy MH, Batra HV. Application of monoclonal antibodies generated against Panton-Valentine Leukocidin (PVL-S) toxin for specific identification of community acquired methicillin resistance Staphylococcus aureus. Microbiol Res 2014; 169:924-30. [PMID: 24935729 DOI: 10.1016/j.micres.2014.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 05/05/2014] [Accepted: 05/09/2014] [Indexed: 10/25/2022]
Abstract
Panton-Valentine Leukocidin (PVL) produced by community acquired methicillin Staphylococcus aureus (CA-MRSA) involved in skin and soft-tissue infections and necrotizing pneumonia comprised of two fractions, namely PVL S and PVL F. In the present study, three monoclonal antibodies designated as MAb1, MAb9 and MAb10 were generated against recombinant PVL-S (35kDa) protein of S. aureus. All the three MAbs specifically reacted to confirm PVL-S positive strains of S. aureus recovered from clinical samples in Western blot analysis. Similarly all the three MAbs did not show any binding to other tested 14 different pathogenic bacteria belonging to other genera and species in Western blot analysis. Furthermore, a simple dot-ELISA method was standardized for the identification of PVL-S toxin containing S. aureus strains. Initially in dot-ELISA, Protein A (Spa) of S. aureus posed background noise problems due to the non-specific binding of antibodies resulting in false positive reactions. With the addition of 10mM diethylpyrocarbonate (DEPC) along with 5% milk in PBS in the blocking step prevented this non-specific binding of Spa to mouse anti-PVL monoclonal antibodies in dot-ELISA. Once standardized, this simple dot-ELISA was evaluated with nine PVL positive strains recovered from food, environmental and clinical samples and the results were compared with PCR assay for the presence of PVL toxin genes and also with Western blot analysis. A 100% correlation was found between dot-ELISA, PCR assay and Western blot analysis. Collectively our results suggest the newly developed simple dot-ELISA system can be of immense help in providing, rapid detection of the PVL toxin containing S. aureus strains at a relatively low cost and will be a valuable tool for the reliable identification of CA-MRSA.
Collapse
Affiliation(s)
- Niveditha Sundar Poojary
- Division of Microbiology, Defence Food Research Laboratory, Siddartha Nagar, Mysore 570011, Karnataka, India.
| | - Shylaja Ramlal
- Division of Microbiology, Defence Food Research Laboratory, Siddartha Nagar, Mysore 570011, Karnataka, India.
| | - Radhika Madan Urs
- Division of Microbiology, Defence Food Research Laboratory, Siddartha Nagar, Mysore 570011, Karnataka, India.
| | | | - Harsh Vardhan Batra
- Division of Microbiology, Defence Food Research Laboratory, Siddartha Nagar, Mysore 570011, Karnataka, India.
| |
Collapse
|
11
|
Jiang M, Yao J, Feng G. Protective effect of DNA vaccine encoding pseudomonas exotoxin A and PcrV against acute pulmonary P. aeruginosa Infection. PLoS One 2014; 9:e96609. [PMID: 24788626 PMCID: PMC4006881 DOI: 10.1371/journal.pone.0096609] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 04/09/2014] [Indexed: 11/18/2022] Open
Abstract
Infections with Pseudomonas aeruginosa have been a long-standing challenge for clinical therapy because of complex pathogenesis and resistance to antibiotics, thus attaching importance to explore effective vaccines for prevention and treatment. In the present study, we constructed a novel DNA vaccine by inserting mutated gene toxAm encoding Pseudomonas Exotoxin A and gene pcrV encoding tip protein of the type III secretion system into respective sites of a eukaryotic plasmid pIRES, named pIRES-toxAm-pcrV, and next evaluated the efficacy of the vaccine in murine acute Pseudomonas pneumonia models. Compared to DNA vaccines encoding single antigen, mice vaccinated with pIRES-toxAm-pcrV elicited higher levels of antigen-specific serum immunoglobulin G (IgG), enhanced splenic cell proliferation and cytokine secretion in response to Pseudomonas aeruginosa antigens, additionally PAO1 challenge in mice airway resulted in reduced bacteria burden and milder pathologic changes in lungs. Besides, it was observed that immunogenicity and protection could be promoted by the CpG ODN 1826 adjuvant. Taken together, it's revealed that recombinant DNA vaccine pIRES-toxAm-pcrV was a potential candidate for immunotherapy of Pseudomonas aeruginosa infection and the CpG ODN 1826 a potent stimulatory adjuvant for DNA vaccination.
Collapse
Affiliation(s)
- Mingzi Jiang
- Department of Respiratory, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jing Yao
- Department of Respiratory, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ganzhu Feng
- Department of Respiratory, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
12
|
Singh AK, Kingston JJ, Murali HS, Batra HV. A recombinant bivalent fusion protein rVE confers active and passive protection against Yersinia enterocolitica infection in mice. Vaccine 2014; 32:1233-9. [DOI: 10.1016/j.vaccine.2014.01.044] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 01/10/2014] [Accepted: 01/15/2014] [Indexed: 01/30/2023]
|
13
|
Bhatnagar S, Naqvi RA, Ali R, Rao DN. Preparation of peptide microspheres using tumor antigen-derived peptides. Methods Mol Biol 2014; 1139:443-452. [PMID: 24619698 DOI: 10.1007/978-1-4939-0345-0_34] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Due to its distinct biological attributes, poly(D,L lactide-co glycolide) (PLGA) is one of the most preferred methods for DNA/protein/peptide encapsulation for therapeutics. Importantly, PLGA acts as an adjuvant for weakly immunogenic antigens and mimics booster responses after a single dose of administration, thereby serving as a single-shot vaccine delivery vehicle. Efficient delivery of antigens to antigen-presenting cells (APC) has been made possible by the use of a PLGA particle-based vaccine delivery system. Also, the plasma half-life of the PLGA-encapsulated vaccine increases as it is protected from degradation, prior to its further release. PLGAs are reported to be catabolized into individual nontoxic units once inside the host and further degraded via normal metabolic pathways. In this chapter, we have described the preparation and characterization of tumor peptide encapsulated PLGA microparticles as a model for controlled-release peptide delivery system.
Collapse
Affiliation(s)
- Santwana Bhatnagar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | | | | | | |
Collapse
|
14
|
Dinglasan RR, Armistead JS, Nyland JF, Jiang X, Mao HQ. Single-dose microparticle delivery of a malaria transmission-blocking vaccine elicits a long-lasting functional antibody response. Curr Mol Med 2013; 13:479-87. [PMID: 23331003 PMCID: PMC3706950 DOI: 10.2174/1566524011313040002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 01/08/2013] [Accepted: 01/12/2013] [Indexed: 12/15/2022]
Abstract
Malaria sexual stage and mosquito transmission-blocking vaccines (SSM-TBV) have recently gained prominence as a necessary tool for malaria eradication. SSM-TBVs are unique in that, with the exception of parasite gametocyte antigens, they primarily target parasite or mosquito midgut surface antigens expressed only inside the mosquito. As such, the primary perceived limitation of SSM-TBVs is that the absence of natural boosting following immunization will limit its efficacy, since the antigens are never presented to the human immune system. An ideal, safe SSM-TBV formulation must overcome this limitation. We provide a focused evaluation of relevant nano-/microparticle technologies that can be applied toward the development of leading SSM-TBV candidates, and data from a proof-of-concept study demonstrating that a single inoculation and controlled release of antigen in mice, can elicit long-lasting protective antibody titers. We conclude by identifying the remaining critical gaps in knowledge and opportunities for moving SSM-TBVs to the field.
Collapse
Affiliation(s)
- R R Dinglasan
- W Harry Feinstone Department of Molecular Microbiology & Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| | | | | | | | | |
Collapse
|
15
|
Ali R, Naqvi RA, Kumar S, Bhat AA, Rao DN. Multiple antigen peptide containing B and T cell epitopes of F1 antigen of Yersinia pestis showed enhanced Th1 immune response in murine model. Scand J Immunol 2013; 77:361-71. [PMID: 23480362 DOI: 10.1111/sji.12042] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 02/19/2013] [Indexed: 12/11/2022]
Abstract
Yersinia pestis is a facultative bacterium that can survive and proliferate inside host macrophages and cause bubonic, pneumonic and systemic infection. Apart from humoral response, cell-mediated protection plays a major role in combating the disease. Fraction 1 capsular antigen (F1-Ag) of Y. pestis has long been exploited as a vaccine candidate. In this study, F1-multiple antigenic peptide (F1-MAP or MAP)-specific cell-mediated and cytokine responses were studied in murine model. MAP consisting of three B and one T cell epitopes of F1-antigen with one palmitoyl residue was synthesized using Fmoc chemistry. Mice were immunized with different formulations of MAP in poly DL-lactide-co-glycolide (PLGA) microspheres. F1-MAP with CpG oligodeoxynucleotide (CpG-ODN) as an adjuvant showed enhanced in vitro T cell proliferation and Th1 (IL-2, IFN-γ and TNF-α) and Th17 (IL-17A) cytokine secretion. Similar formulation also showed significantly higher numbers of cytokine (IL-2, IFN-γ)-secreting cells. Moreover, F1-MAP with CpG formulation showed significantly high (P < 0.001) percentage of CD4(+) IFN-γ(+) cells as compared to CD8(+) IFN-γ(+) cells, and also more (CD4- IFN-γ)(+) cells secrete perforin and granzyme as compared to (CD8- IFN-γ)(+) showing Th1 response. Thus, the study highlights the importance of Th1 cytokine and existence of CD4(+) and CD8(+) immune response. This study proposes a new perspective for the development of vaccination strategies for Y. pestis that trigger T cell immune response.
Collapse
Affiliation(s)
- R Ali
- Department of Biochemistry, All India Institute of Medical Sciences AIIMS, New Delhi-110029, India
| | | | | | | | | |
Collapse
|
16
|
Ali R, Kumar S, Naqvi RA, Sheikh IA, Rao DN. Multiple antigen peptide consisting of B- and T-cell epitopes of F1 antigen of Y. pestis showed enhanced humoral and mucosal immune response in different strains of mice. Int Immunopharmacol 2012; 15:97-105. [PMID: 23174507 DOI: 10.1016/j.intimp.2012.10.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 10/29/2012] [Accepted: 10/31/2012] [Indexed: 01/08/2023]
Abstract
Yersinia pestis is a causative agent of plague. F1 and V antigen based vaccines have shown remarkable protection in experimental animals. In order to develop epitope based immunogen, three B and one T-cell epitopes of F1 antigen with palmitate residue at amino terminal were assembled on a lysine backbone as multiple antigen peptide (MAP or F1-MAP). MAP was characterized by SDS-PAGE, immunoblot and immunoreactivity with anti F1 sera. MAP was entrapped in PLGA (polylactide-co-glycolide) microparticles and humoral, mucosal immune responses were studied after intranasal immunization with/without CpG ODN 1826 (CpG)/murabutide in different strains of mice. Serum and mucosal washes were measured for MAP specific IgG, IgA, sIgA and IgG subclasses in three strains of mice. F1-MAP showed high serum antibody and mucosal IgG and IgA peak antibody titers. MAP with CpG showed significantly high (p<0.001) peak antibody titer ranging from 102,400 to 204,800 for IgG and 6400 to 12,800 for IgA. High mucosal sIgA and its secretary component detection confirmed generation of mucosal response in intestinal and lung washes. MAP antisera also showed significant immunoreactivity with individual peptides. Moreover, antibody specific activity (IgG, IgA and sIgA) positively correlates with peak antibody titers. Predominantly IgG2a/IgG2b subclass was observed with CpG formulation but in other formulation a mixed IgG1 and IgG2a response was observed. The present study highlights the importance of multiple antigen peptide approach of F1-antigen with CpG as an alternative approach for subunit vaccine.
Collapse
Affiliation(s)
- Riyasat Ali
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | | | | | | | | |
Collapse
|
17
|
Intranasal vaccination with murabutide enhances humoral and mucosal immune responses to a virus-like particle vaccine. PLoS One 2012; 7:e41529. [PMID: 22855691 PMCID: PMC3405106 DOI: 10.1371/journal.pone.0041529] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 06/27/2012] [Indexed: 01/12/2023] Open
Abstract
Murabutide (MB) is a synthetic immunomodulator recognized by the nucleotide-binding oligomerization domain-containing protein 2 (NOD2) receptor on mammalian cells. MB has previously been approved for testing in multiple human clinical trials to determine its value as an antiviral therapeutic, and as an adjuvant for injected vaccines. We have found a new use for this immunomodulator; it functions as a mucosal adjuvant that enhances immunogenicity of virus-like particles (VLP) administered intranasally. MB enhanced Norwalk virus (NV) VLP-specific IgG systemically and IgA production at distal mucosal sites following intranasal (IN) vaccination. A dose escalation study identified 100 µg as the optimal MB dosage in mice, based on the magnitude of VLP-specific IgG, IgG1, IgG2a and IgA production in serum and VLP-specific IgA production at distal mucosal sites. IN vaccination using VLP with MB was compared to IN delivery VLP with cholera toxin (CT) or gardiquimod (GARD) and to parenteral VLP delivery with alum; the MB groups were equivalent to CT and GARD and superior to alum in inducing mucosal immune responses and stimulated equivalent systemic VLP-specific antibodies. These data support the further testing of MB as a potent mucosal adjuvant for inducing robust and durable antibody responses to non-replicating subunit vaccines.
Collapse
|
18
|
Shreewastav RK, Ali R, Uppada JB, Rao DN. Cell-mediated immune response to epitopic MAP (multiple antigen peptide) construct of LcrV antigen of Yersinia pestis in murine model. Cell Immunol 2012; 278:55-62. [PMID: 23121976 DOI: 10.1016/j.cellimm.2012.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 06/20/2012] [Accepted: 07/07/2012] [Indexed: 12/14/2022]
Abstract
Yersinia pestis is the causative agent of plague. Cellular immunity seems to play an important role in defense against this disease. The subunit vaccine based on V (Lcr V) antigen has been proved to be immunogenic in animals and in humans. The multiple antigen peptide (MAP), incorporating all the relevant B and T cell epitopes is highly immunogenic in mice through intranasal route of immunization in PLGA particles containing CpG-ODN as an immunoadjuvant inducing humoral and mucosal immune response. In the present study, cell-mediated immune response using same MAP was studied in murine model. Primary and memory T cell responses were studied in outbred and inbred mice immunized intranasally with MAP in the presence of two immunoadjuvants (Murabutide and CpG-ODN). All the three compartments (Spleen, Lamina propria and Peyer's patches) of the lymphoid system showed increased lymphoproliferative response. Highest lymphoproliferative response was observed especially with CpG-ODN. Cytokine profile in the culture supernatant showed highest Th(1) and Th(17) levels. FACS analysis showed expansion of both CD4(+) and CD8(+) T cells producing gamma-interferon, perforin and granzyme-B with major contribution from CD4(+) T cells.
Collapse
|