1
|
Sanches RCO, Tiwari S, Ferreira LCG, Oliveira FM, Lopes MD, Passos MJF, Maia EHB, Taranto AG, Kato R, Azevedo VAC, Lopes DO. Immunoinformatics Design of Multi-Epitope Peptide-Based Vaccine Against Schistosoma mansoni Using Transmembrane Proteins as a Target. Front Immunol 2021; 12:621706. [PMID: 33737928 PMCID: PMC7961083 DOI: 10.3389/fimmu.2021.621706] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 02/08/2021] [Indexed: 12/17/2022] Open
Abstract
Schistosomiasis remains a serious health issue nowadays for an estimated one billion people in 79 countries around the world. Great efforts have been made to identify good vaccine candidates during the last decades, but only three molecules reached clinical trials so far. The reverse vaccinology approach has become an attractive option for vaccine design, especially regarding parasites like Schistosoma spp. that present limitations for culture maintenance. This strategy also has prompted the construction of multi-epitope based vaccines, with great immunological foreseen properties as well as being less prone to contamination, autoimmunity, and allergenic responses. Therefore, in this study we applied a robust immunoinformatics approach, targeting S. mansoni transmembrane proteins, in order to construct a chimeric antigen. Initially, the search for all hypothetical transmembrane proteins in GeneDB provided a total of 584 sequences. Using the PSORT II and CCTOP servers we reduced this to 37 plasma membrane proteins, from which extracellular domains were used for epitope prediction. Nineteen common MHC-I and MHC-II binding epitopes, from eight proteins, comprised the final multi-epitope construct, along with suitable adjuvants. The final chimeric multi-epitope vaccine was predicted as prone to induce B-cell and IFN-γ based immunity, as well as presented itself as stable and non-allergenic molecule. Finally, molecular docking and molecular dynamics foresee stable interactions between the putative antigen and the immune receptor TLR 4. Our results indicate that the multi-epitope vaccine might stimulate humoral and cellular immune responses and could be a potential vaccine candidate against schistosomiasis.
Collapse
Affiliation(s)
- Rodrigo C. O. Sanches
- Laboratório de Biologia Molecular, Universidade Federal de São João del-Rei, Divinópolis, Brazil
| | - Sandeep Tiwari
- Programa de Pós-Graduação em Bioinformática, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Laís C. G. Ferreira
- Laboratório de Biologia Molecular, Universidade Federal de São João del-Rei, Divinópolis, Brazil
| | - Flávio M. Oliveira
- Laboratório de Biologia Molecular, Universidade Federal de São João del-Rei, Divinópolis, Brazil
| | - Marcelo D. Lopes
- Laboratório de Biologia Molecular, Universidade Federal de São João del-Rei, Divinópolis, Brazil
| | - Maria J. F. Passos
- Laboratório de Biologia Molecular, Universidade Federal de São João del-Rei, Divinópolis, Brazil
| | - Eduardo H. B. Maia
- Laboratório de Química Farmacêutica Medicinal, Universidade Federal de São João del-Rei, Divinópolis, Brazil
- Centro Federal de Educação Tecnológica de Minas Gerais (CEFET-MG), Divinópolis, Brazil
| | - Alex G. Taranto
- Laboratório de Química Farmacêutica Medicinal, Universidade Federal de São João del-Rei, Divinópolis, Brazil
| | - Rodrigo Kato
- Programa de Pós-Graduação em Bioinformática, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Vasco A. C. Azevedo
- Programa de Pós-Graduação em Bioinformática, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Debora O. Lopes
- Laboratório de Biologia Molecular, Universidade Federal de São João del-Rei, Divinópolis, Brazil
| |
Collapse
|
2
|
Chen L, Zhang D, Zhang W, Zhu Y, Hou M, Yang B, Xu Z, Ji M, Wu G. Absence of Batf3 results in reduced liver pathology in mice infected with Schistosoma japonicum. Parasit Vectors 2017. [PMID: 28646891 PMCID: PMC5483257 DOI: 10.1186/s13071-017-2250-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background The involvement of CD8+T cells in schistosomiasis is being increasingly appreciated, but the underlying mechanism is not well defined. Results In this study, we showed that the absence of Batf3 alleviated liver damage in Batf3−/− mice infected with S. japonicum. We found alleviated liver granulomatous inflammation in Batf3−/− mice with schistosomiasis japonica could not be attributed to the difference in schistosome egg or worm burden. The stronger Tc1 cell responses observed in Batf3−/− mice suggested that the deletion of Batf3 resulted in more activation of CD8+T cells unexpectedly during the natural infection of schistosomes. We detected a small amount of CD8α+ DCs in the spleen of Batf3−/− mice at 9w post-infection. This small amount of newly generated CD8α+ DCs might contribute to enhanced activation of CD8+T cells via cross-presentation and activation which then attenuate hepatic pathological damage found in Batf3−/− mice. Conclusions Our study provides evidence that Batf3 is associated with the immunoregulation of the liver granuloma formation, which may confer a new options for schistosomiasis treatment.
Collapse
Affiliation(s)
- Lin Chen
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China. .,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China.
| | - Donghui Zhang
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China.,School of International Education, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenyue Zhang
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Yuxiao Zhu
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China
| | - Min Hou
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China
| | - Bingya Yang
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China
| | - Zhipeng Xu
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China
| | - Minjun Ji
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China
| | - Guanling Wu
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China
| |
Collapse
|
3
|
Chen L, Chen Y, Zhang D, Hou M, Yang B, Zhang F, Zhang W, Luo X, Ji M, Wu G. Protection and immunological study on two tetraspanin-derived vaccine candidates against schistosomiasis japonicum. Parasite Immunol 2016; 38:589-98. [PMID: 27189226 DOI: 10.1111/pim.12338] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 05/12/2016] [Indexed: 12/29/2022]
Abstract
Tetraspanins (TSPs) are proteins found on the surface of helminth parasites of the genus Schistosoma and are regarded as potentially protective antigens. The large extracellular loop of Schistosoma mansoni tetraspanin-2, Sm-TSP-2, when fused to a thioredoxin partner and formulated with Freund's adjuvants, has been shown to be an efficacious vaccine against murine schistosomiasis. It is well recognized that CD4(+) T-cell-dependent immunity might play an important role against schistosomes; however, the contribution of CD8(+) T cells against multicellular pathogen is still uncertain. The exogenous protein-pulsed dendritic cells (DCs) can easily activate CD4(+) T cells response, while CD8(+) T cells response was relatively difficult to be induced. In this study, we evaluated the immunogenicity of TSP2HD antigen (hydrophilic domain of the S. japonicum tetraspanin-2) and TAT (the protein transduction domain of HIV-1)-coupled TSP2HD protein. As TAT-fused protein could promote major histocompatibility complex class I-dependent antigen presentation in vitro, TAT-TSP2HD-pulsed DCs induced stronger proliferation of schistosome-specific CD8(+) T cells compared with DCs incubated with TSP2HD alone. Vaccination with TAT-TSP2HD-pulsed DCs in vivo could improve disease outcome in S. japonicum-infected mice and was slightly superior to vaccination with DCs treated with TSP2HD. In summary, these data showed that TAT fusion proteins could help activate CD8(+) cells and Th1 cells and provide part protection against schistosome.
Collapse
Affiliation(s)
- L Chen
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China
| | - Y Chen
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - D Zhang
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China
| | - M Hou
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China
| | - B Yang
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China
| | - F Zhang
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - W Zhang
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - X Luo
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - M Ji
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China. .,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China.
| | - G Wu
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China
| |
Collapse
|
4
|
Zhang W, Luo X, Zhang F, Zhu Y, Yang B, Hou M, Xu Z, Yu C, Chen Y, Chen L, Ji M. SjTat-TPI facilitates adaptive T-cell responses and reduces hepatic pathology during Schistosoma japonicum infection in BALB/c mice. Parasit Vectors 2015; 8:664. [PMID: 26714844 PMCID: PMC4696208 DOI: 10.1186/s13071-015-1275-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 12/18/2015] [Indexed: 12/18/2022] Open
Abstract
Background Schistosomiasis is a kind of parasitic zoonoses which causes serious damage to public health and social development. China is one of the countries most affected by Schistosoma japonicum and an effective vaccine is still needed. In this study, we adopted Tat-mediated protein transduction technology to investigate the impact of different antigen presented approaches on host’s immune response and the potential protection against Schistosoma japonicum infection. Results We successfully constructed the recombinant S. japonicum triosephosphate isomerase, Tat-TPI, as a vaccine candidate. Whether injected with Tat-TPI in foot pad or vaccinated with Tat-TPI in the back subcutaneously for three times, the draining popliteal lymph nodes and spleen both developed a stronger CD8+T response (Tc1) in mice. Not only that, but it also helped CD4+T cells to produce more IFN-γ than TPI immunisation. In addition, it could boost IgG production, especially IgG1 subclass. Most importantly, Tat-TPI immunisation led to the significant smaller area of a single egg granuloma in the livers as compared with TPI-vaccinated or control groups. However, the anti-infection efficiency induced by Tat-TPI was still restricted. Conclusion This study indicated that immunisation with Tat-fused TPI could contribute to enhance CD4+T-cell response and decrease hepatic egg granulomatous area after S. japonicum infection though it did not achieve our expected protection against Schistosoma japonicum infection. The optimal vaccine strategy warrants further research.
Collapse
Affiliation(s)
- Wenyue Zhang
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| | - Xiaofeng Luo
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| | - Fan Zhang
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| | - Yuxiao Zhu
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| | - Bingya Yang
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China. .,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, 210029, China.
| | - Min Hou
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| | - Zhipeng Xu
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China. .,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, 210029, China.
| | - Chuanxin Yu
- Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu, 214064, China.
| | - Yingying Chen
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| | - Lin Chen
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China. .,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, 210029, China.
| | - Minjun Ji
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China. .,Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
5
|
Fonseca CT, Oliveira SC, Alves CC. Eliminating Schistosomes through Vaccination: What are the Best Immune Weapons? Front Immunol 2015; 6:95. [PMID: 25806033 PMCID: PMC4353369 DOI: 10.3389/fimmu.2015.00095] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 02/18/2015] [Indexed: 12/24/2022] Open
Abstract
The successful development of vaccines depends on the knowledge of the immunological mechanisms associated with the elimination of the pathogen. In the case of schistosomes, its complex life cycle and the mechanisms developed to evade host immune system, turns the development of a vaccine against the disease into a very difficult task. Identifying the immunological effector mechanisms involved in parasite attrition and the major targets for its response is a key step to formulate an effective vaccine. Recent studies have described some promising antigens to compose a subunit vaccine and have pointed to some immune factors that play a role in parasite elimination. Here, we review the immune components and effector mechanisms associated with the protective immunity induced by those vaccine candidates and the lessons we have learned from the studies of the acquired resistance to infection in humans. We will also discuss the immune factors that correlate with protection and therefore could help to evaluate those vaccine formulations in clinical trials.
Collapse
Affiliation(s)
- Cristina Toscano Fonseca
- Laboratório de Esquistossomose do Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz , Belo Horizonte , Brazil ; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação, Universidade Federal de Minas Gerais , Belo Horizonte , Brazil
| | - Sergio Costa Oliveira
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação, Universidade Federal de Minas Gerais , Belo Horizonte , Brazil ; Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais , Belo Horizonte , Brazil
| | - Clarice Carvalho Alves
- Laboratório de Esquistossomose do Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz , Belo Horizonte , Brazil
| |
Collapse
|
6
|
Upregulated expression of cytotoxicity-related genes in IFN-γ knockout mice with Schistosoma japonicum infection. J Biomed Biotechnol 2011; 2011:864945. [PMID: 21976969 PMCID: PMC3184444 DOI: 10.1155/2011/864945] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Accepted: 07/28/2011] [Indexed: 11/17/2022] Open
Abstract
It is well accepted that IFN-γ is important to the development of acquired resistance against murine schistosomiasis. However, the in vivo role of this immunoregulatory cytokine in helminth infection needs to be further investigated. In this study, parasite burden and host immune response were observed in IFN-γ knockout mice (IFNg KO) infected with Schistosoma japonicum for 6 weeks. The results suggested that deficiency in IFN-γ led to decreased egg burden in mice, with low schistosome-specific IgG antibody response and enhanced activation of T cells during acute infection. Microarray and qRT-PCR data analyses showed significant upregulation of some cytotoxicity-related genes, including those from the granzyme family, tumor necrosis factor, Fas Ligand, and chemokines, in the spleen cells of IFNg KO mice. Furthermore, CD8+ cells instead of NK cells of IFNg KO mice exhibited increased transcription of cytotoxic genes compared with WT mice. Additionally, Schistosoma japonicum-specific egg antigen immunization also could activate CD8+ T cells to upregulate the expression of cytotoxic genes in IFNg KO mice. Our data suggest that IFN-γ is not always a positive regulator of immune responses. In certain situations, the disruption of IFN-γ signaling may up-regulate the cytotoxic T-cell-mediated immune responses to the parasite.
Collapse
|
7
|
Wang X, Zhang L, Chi Y, Hoellwarth J, Zhou S, Wen X, He L, Liu F, Wu C, Su C. The nature and combination of subunits used in epitope-based Schistosoma japonicum vaccine formulations affect their efficacy. Parasit Vectors 2010; 3:109. [PMID: 21087526 PMCID: PMC3136145 DOI: 10.1186/1756-3305-3-109] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 11/19/2010] [Indexed: 01/07/2023] Open
Abstract
Background Schistosomiasis remains a major public health problem in endemic countries and is caused by infections with any one of three primary schistosome species. Although there are no vaccines available to date, this strategy appears feasible since natural immunity develops in individuals suffering from repeated infection during a lifetime. Since vaccinations resulting in both Th1- and Th2-type responses have been shown to contribute to protective immunity, a vaccine formulation with the capacity for stimulating multiple arms of the immune response will likely be the most effective. Previously we developed partially protective, single Th- and B cell-epitope-based peptide-DNA dual vaccines (PDDV) (T3-PDDV and B3-PDDV, respectively) capable of eliciting immune responses against the Schistosoma japonicum 22.6 kDa tegument antigen (Sj22.6) and a 62 kDa fragment of myosin (Sj62), respectively. Results In this study, we developed PDDV cocktails containing multiple epitopes of S. japonicum from Sj22.6, Sj62 and Sj97 antigens by predicting cytotoxic, helper, and B-cell epitopes, and evaluated vaccine potential in vivo. Results showed that mice immunized with a single-epitope PDDV elicited either Tc, Th, or B cell responses, respectively, and mice immunized with either the T3- or B3- single-epitope PDDV formulation were partially protected against infection. However, mice immunized with a multicomponent (3 PDDV components) formulation elicited variable immune responses that were less immunoprotective than single-epitope PDDV formulations. Conclusions Our data show that combining these different antigens did not result in a more effective vaccine formulation when compared to each component administered individually, and further suggest that immune interference resulting from immunizations with antigenically distinct vaccine targets may be an important consideration in the development of multicomponent vaccine preparations.
Collapse
Affiliation(s)
- Xuefeng Wang
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 210029, PR China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Tian F, Lin D, Wu J, Gao Y, Zhang D, Ji M, Wu G. Immune events associated with high level protection against Schistosoma japonicum infection in pigs immunized with UV-attenuated cercariae. PLoS One 2010; 5:e13408. [PMID: 20976218 PMCID: PMC2955526 DOI: 10.1371/journal.pone.0013408] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Accepted: 09/22/2010] [Indexed: 12/28/2022] Open
Abstract
Background The vaccination of radiation-attenuated Schistosoma japonicum cercariae can induce effective protection in artiodactyl, but the immune events related to protective immunity are not fully understood. To provide a paradigm for a human recombinant antigen vaccine, we have undertaken a vaccination and challenge experiment in pigs, which was recognized as an appropriate animal model in this type of study because of their similarity to human in immunology, and investigated the relative immune events induced by the radiation-attenuated S. japonicum cercariae. Methods and Findings We found that pigs immunized once with 400 µw UV-irradiated cercariae exhibited 63.84% and 71.82% reductions in worm burden and hepatic eggs respectively. Protective immunity in vaccinated pigs was associated with high level productions of IgM, total IgG, IgG1 and IgG2; IgG2 was significantly increased in the acute infection. IFN-γ levels could be elicited by immunization. At week 6 post-infection, IFN-γ, IL-4 and IL-10 levels also showed a dramatic rise synchronously in vaccinated pigs. Moreover, the granzyme b, nk-lysin, ifnγ, il4 and il10 mRNA levels in early skin-draining lymph nodes of immunized pigs were higher than those in pigs with non-irradiated cercariae infection. In addition, cytotoxicity-related genes in the mesenteric lymph nodes were significantly upregulated in vaccinated pigs in the acute infection. Conclusion/Significance Our results demonstrated that IFN-γ and IgG2 antibody production, as well as genes related to cytotoxicity are associated with the high level protection induced by UV-irradiated Schistosoma japonicum vaccine. These findings indicated that optimal vaccination against S. japonicum required the induction of IFN-γ, IgG2 antibody related to Th1 responses and cytotoxicity effect.
Collapse
Affiliation(s)
- Fang Tian
- Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Pathogen Biology and Immunology, Yangzhou University Medical College, Yangzhou, Jiangsu, China
| | - Dandan Lin
- Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangxi Provincial Institute of Parasitic Diseases, Nanchang, Jiangxi, China
| | - Jingjiao Wu
- Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yanan Gao
- Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Donghui Zhang
- Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China
| | - Minjun Ji
- Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China
- * E-mail:
| | - Guanling Wu
- Department of Pathogen Biology and Immunology, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Province Key Laboratory of Modern Pathogen Biology, Nanjing, Jiangsu, China
| |
Collapse
|
9
|
DNA vaccination by electroporation and boosting with recombinant proteins enhances the efficacy of DNA vaccines for Schistosomiasis japonica. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2009; 16:1796-803. [PMID: 19812258 DOI: 10.1128/cvi.00231-09] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Schistosomiasis japonica is an endemic, zoonotic disease of major public health importance in China. Control programs combining chemotherapy and snail killing have not been able to block transmission of infection in lakes and marsh regions. Vaccination is needed as a complementary approach to the ongoing control programs. In the present study, we wanted to determine if the efficacies of DNA vaccines encoding the 23-kDa tetraspanin membrane protein (SjC23), triose phosphate isomerase (SjCTPI), and sixfold-repeated genes of the complementarity determining region 3 (CDR3) in the H chain of NP30 could be enhanced by boosting via electroporation in vivo and/or with cocktail protein vaccines. Mice vaccinated with cocktail DNA vaccines showed a significant worm reduction of 32.88% (P < 0.01) and egg reduction of 36.20% (P < 0.01). Vaccine efficacy was enhanced when animals were boosted with cocktail protein vaccines; adult worm and liver egg burdens were reduced 45.35% and 48.54%, respectively. Nearly identical results were obtained in mice boosted by electroporation in vivo, with adult worm and egg burdens reduced by 45.00% and 50.88%, respectively. The addition of a protein vaccine boost to this regimen further elevated efficacy to approximately 60% for adult worm burden and greater than 60% for liver egg reduction. The levels of interleukin-2, gamma interferon, and the ratios of immunoglobulin G2a (IgG2a)/IgG1 clearly showed that cocktail DNA vaccines induced CD4(+) Th1-type responses. Boosting via either electroporation or with recombinant proteins significantly increased associated immune responses over those seen in mice vaccinated solely with DNA vaccines. Thus, schistosome DNA vaccine efficacy was significantly enhanced via boosting by electroporation in vivo and/or cocktail protein vaccines.
Collapse
|
10
|
Wang X, Jin H, Du X, Cai C, Yu Y, Zhao G, Su B, Huang S, Hu Y, Luo D, She R, Luo X, Zeng X, Yi X, Wang B. The protective efficacy against Schistosoma japonicum infection by immunization with DNA vaccine and levamisole as adjuvant in mice. Vaccine 2008; 26:1832-45. [PMID: 18329762 DOI: 10.1016/j.vaccine.2008.01.056] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2007] [Revised: 01/16/2008] [Accepted: 01/30/2008] [Indexed: 11/28/2022]
Abstract
Levamisole (LMS) as an adjuvant enhances cell-mediated immunity in DNA vaccination; we investigated the efficacy and liver immunopathology alleviation of a DNA vaccine, VR1012-SjGST-32, in a LMS formulation in the murine challenge model. Compared to controls, the VR1012-SjGST-32 plus LMS can reduce worm and egg burdens, as well as, immunopathological complications associated chronic inflammation significantly in liver, which were apparently associated with Th1-type response. Together, these results suggest that the LMS as a potential Schistosome DNA vaccine adjuvant can enhance both worm killing and disease prevention, which is possibly mediated through the induction of a strong Th1-dominant environment in immunized mice.
Collapse
Affiliation(s)
- Xiao Wang
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, 2 Yuanmingyuan Xi Road, Beijing 100094, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
El Ridi R, Salem R, Wagih A, Mahana N, El Demellawy M, Tallima H. Influence of interleukin-2 and interferon-gamma in murine schistosomiasis. Cytokine 2006; 33:281-8. [PMID: 16564704 DOI: 10.1016/j.cyto.2006.01.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2005] [Revised: 10/16/2005] [Accepted: 01/18/2006] [Indexed: 10/24/2022]
Abstract
Schistosoma mansoni-infected mice were administered at the time of parasite residency in the lung with recombinant murine interleukin (IL)-2 or interferon-gamma (IFN-gamma), to evaluate the impact of cytokines in host responses to primary schistosomiasis. S. mansoni lung-stage schistosomula did not affect plasma lipids levels in BALB/c, while elicited significant (p<0.05) increase in free fatty acids (FA) and decrease in cholesterol plasma levels in C57BL/6 and CD1 mice, and stimulated expression of mRNA for Th2 cytokines in BALB/c and Th1 cytokines in C57BL/6 and CD1 mice. Production of specific antibodies was negligible in the 3 strains. Interleukin-2 treatment elicited significant (p<0.001) decrease in triglycerides (TG) in CD1, and decrease in TG and cholesterol plasma levels and down-regulation of TNF-alpha mRNA expression in C57BL/6 mice. Induction of type 2 cytokines and/or IFN-gamma mRNA expression did not lead to increase in percentage of specific antibody responders in any mouse strain. Exogenous IL-2-related reduction in cholesterol plasma levels and TNF-alpha mRNA expression in C57BL/6 mice was associated with significant (p<0.05) decrease in adult worm recovery and egg count. Treatment with IFN-gamma elicited significant (p<0.05) free FA plasma levels increase in BALB/c and C57BL/6 and decrease in CD1 mice. Expression of type 2 cytokines mRNA was stimulated in BALB/c and CD1 mice, yet was not accompanied with increase in humoral responses. Exogenous IFN-gamma-related reduction in free FA plasma levels and IFN-gamma mRNA response, and up-regulation of TNF-alpha mRNA expression in CD1 mice were associated with significant increase in adult worm burden and egg load. The data were discussed in an attempt to define host factors predictive of resistance to schistosome infection.
Collapse
Affiliation(s)
- Rashika El Ridi
- Zoology Department, Faculty of Science, Cairo University, Cairo 12613, Egypt.
| | | | | | | | | | | |
Collapse
|
12
|
Liu P, Hu YY, Liu C, Zhu DY, Xue HM, Xu ZQ, Xu LM, Liu CH, Gu HT, Zhang ZQ. Clinical observation of salvianolic acid B in treatment of liver fibrosis in chronic hepatitis B. World J Gastroenterol 2002; 8:679-85. [PMID: 12174378 PMCID: PMC4656320 DOI: 10.3748/wjg.v8.i4.679] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2001] [Revised: 12/12/2001] [Accepted: 12/20/2001] [Indexed: 02/06/2023] Open
Abstract
AIM To evaluate the clinical efficacy of salvianolic acid B (SA-B) on liver fibrosis in chronic hepatitis B. METHODS Sixty patients with definite diagnosis of liver fibrosis with hepatitis B were included in the trial. Interferon-gamma (IFN-gamma) was used as control drug. The patients took orally SA-B tablets or received muscular injection of IFN-gamma in the double blind randomized test. The complete course lasted 6 months. The histological changes of liver biopsy specimen before and after the treatment were the main evidence in evaluation, in combination with the results of contents of serum HA, LN, IV-C, P-III-P, liver ultrasound imaging, and symptoms and signs. RESULTS Reverse rate of fibrotic stage was 36.67 % in SA-B group and 30.0 % in IFN-gamma group. Inflammatory alleviating rate was 40.0 % in SA-B group and 36.67 % in IFN-gamma group. The average content of HA and IV-C was significantly lower than that before treatment. The abnormal rate also decreased remarkably. Overall analysis of 4 serological fibrotic markers showed significant improvement in SA-B group as compared with the IFN-gamma group. Score of liver ultrasound imaging was lower in SA-B group than in IFN-gamma group (HA 36.7 % vs 80 %, IV-C 3.3 % vs 23.2 %). Before the treatment, ALT AST activity and total bilirubin content of patients who had regression of fibrosis after oral administration of SA-B, were significantly lower than those of patients who had aggravation of fibrosis after oral administration of SA-B. IFN-gamma showed certain side effects (fever and transient decrease of leukocytes, occurrence rates were 50 % and 3.23 %), but SA-B showed no side effects. CONCLUSION SA-B could effectively reverse liver fibrosis in chronic hepatitis B. SA-B was better than IFN-gamma in reduction of serum HA content, overall decrease of 4 serum fibrotic markers, and decrease of ultrasound imaging score. Liver fibrosis in chronic hepatitis B with slight liver injury was more suitable to SA-B in anti-fibrotic treatment. SA-B showed no obvious side effects.
Collapse
Affiliation(s)
- Ping Liu
- Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Verity CK, McManus DP, Brindley PJ. Cellular responses to Schistosoma japonicum cathepsin D aspartic protease. Parasite Immunol 2002; 24:363-7. [PMID: 12164822 DOI: 10.1046/j.1365-3024.2002.00475.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Lymphocyte proliferation and cytokine production were measured in groups of mice vaccinated (but not subsequently challenge infected) with recombinant forms of Schistosoma japonicum cathepsin D aspartic protease, rSjASP1 (expressed in bacteria; enzymatically inactive) and rSjASP2 (expressed in insect cells; enzymatically active). Both forms of the schistosome enzyme induced significant proliferation of splenocytes recovered from vaccinated mice, and expression of interferon (IFN)-gamma, interleukin (IL)-4 and IL-10 mRNA in these cells was detected using reverse transcriptase-polymerase chain reaction. Secretion of IFN-gamma, IL-4 and IL-10 by splenocytes from vaccinated mice was confirmed and quantified using enzyme-linked immunosorbent assay. IFN-gamma was the most abundant cytokine produced, followed by IL-4 and IL-10 in rank order. These findings indicated that vaccination of mice with the schistosome protease induces a mixed Th1/Th2 cytokine response, which may explain the modest level of protection after challenge infection in cathepsin D-vaccinated mice, reported previously.
Collapse
Affiliation(s)
- Christiana K Verity
- Division of Infectious Diseases and Immunology and CRC for Vaccine Technology, The Queensland Institute of Medical Research, Royal Brisbane Hospital, Queensland, Australia
| | | | | |
Collapse
|
14
|
Angyalosi G, Neveu R, Wolowczuk I, Delanoye A, Herno J, Auriault C, Pancré V. HLA class II polymorphism influences onset and severity of pathology in Schistosoma mansoni-infected transgenic mice. Infect Immun 2001; 69:5874-82. [PMID: 11500466 PMCID: PMC98706 DOI: 10.1128/iai.69.9.5874-5882.2001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Genetic factors that might influence susceptibility or resistance in naive individuals and early-stage pathology in schistosomiasis are difficult to study in clinical trials, since in areas where the disease is endemic the first contact with the parasite occurs most often at very early ages. Therefore, four strains (DR1.Abeta degrees, DR2.Abeta degrees, DQ8.Abeta degrees, and DQ6.Abeta degrees ) of major histocompatibility complex class II-deficient mice (Abeta degrees ), transgenic for different HLA alleles, have been used to evaluate the potential role of HLA class II polymorphism in the onset of the infection by Schistosoma mansoni. The survival rates and parasitological and immunological parameters after infection were evaluated and compared against the control values obtained with Abeta degrees mice. All four mouse strains used in this study were able to generate a specific immune response against S. mansoni antigens (cytokine production and antibody production). However, only mice expressing DR alleles survived until the chronic stage of the infection and were able to mount protective granulomatous response avoiding hepatic damage, presenting predominant gamma interferon production. In contrast, strains expressing DQ alleles revealed an impairment in generating effective granulomas, resulting in earlier death, which was associated with an impaired hepatic granulomatous response and liquefactic necrosis, reflecting the influence of HLA polymorphism in the establishment of protective response in the early stage of infection.
Collapse
Affiliation(s)
- G Angyalosi
- Laboratoire d'Immunopathologie Cellulaire des Maladies Infectieuses, UMR 8527, Institut de Biologie, F-59021 Lille, France.
| | | | | | | | | | | | | |
Collapse
|
15
|
Dupré L, Kremer L, Wolowczuk I, Riveau G, Capron A, Locht C. Immunostimulatory effect of IL-18-encoding plasmid in DNA vaccination against murine Schistosoma mansoni infection. Vaccine 2001; 19:1373-80. [PMID: 11163659 DOI: 10.1016/s0264-410x(00)00363-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
In vivo delivery of DNA encoding antigens is a simple tool to induce immune responses against pathogens. This approach to vaccination also offers the possibility to codeliver plasmids encoding immunomodulatory molecules in order to drive immune responses towards optimal protective effects. In the murine model of Schistosoma mansoni infection, vaccination inducing a Th1 profile has been shown to be protective. In this study, we used a plasmid encoding the Th1-promoting cytokine IL-18, since we observed that percutaneous infection of Balb/c mice strongly induced the production of IL-18 mRNA in the skin. Intradermal injection of the IL-18-encoding plasmid prior to infection did not interfere with parasite migration through the skin although it led to a local and transient cellular infiltration. When the IL-18-encoding plasmid was codelivered with a S. mansoni glutathione S-transferase (Sm28GST)-encoding plasmid, a 30-fold increase of antigen-specific IFN-gamma secretion by spleen cells was observed in comparison to spleen cells from mice that had received only the Sm28GST-encoding plasmid. This immunostimulatory effect was related to a significant protective effect (28% reduction in egg laying and 23% reduction in worm burden) which was attributed to a cooperative effect between both plasmids. Therefore, this study shows that codelivery of an IL-18-encoding plasmid with an antigen-encoding plasmid can stimulate specific cellular responses and induce protective effects against S. mansoni infection.
Collapse
Affiliation(s)
- L Dupré
- Laboratoire des Relations Hôtes-Parasite et Stratégies Vaccinales, INSERM U 167, Institut Pasteur de Lille, F-59019 Cedex, Lille, France
| | | | | | | | | | | |
Collapse
|
16
|
Pancré V, Delacre M, Herno J, Auriault C. Schistosomal egg antigen-responsive CD8 T-cell population in Schistosoma mansoni-infected BALB/c mice. Immunology 1999; 98:525-34. [PMID: 10594684 PMCID: PMC2326969 DOI: 10.1046/j.1365-2567.1999.00887.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We demonstrated here that schistosomal egg antigen (SEA) is able to stimulate an antigen-specific, cytotoxic CD8+ T-cell response in mice. Indeed, a single i.p. immunization with SEA resulted in the in vivo induction of significant cytotoxic T lymphocyte (CTL) activity in the spleen within 20 days. Effector cells were classic class I major histocompatibility complex (MHC)-restricted CD8+ lymphocytes producing interferon-gamma (IFN-gamma) and interleukin-2 (IL-2), suggesting a type 1 response to SEA. We therefore investigated the relevance of these observations in the context of the Schistosoma mansoni parasite infection. CTL activity against SEA-pulsed target cells was evidenced throughout the infection after in vitro stimulation of recovered splenic cells with SEA demonstrating that SEA-specific CD8+ T cells with cytotoxic potentialities are present during infection. This activity was strongly increased after immunization of mice with SEA like the production of IFN-gamma in the sera. A marked reduction in the number of granulomas and of fibrosis with the presence of cells producing IFN-gamma in the liver was also observed leading to the survival of SEA-immunized mice.
Collapse
Affiliation(s)
- V Pancré
- Laboratoire dImmunopathologie cellulaire des maladies infectieuses, Lille, France
| | | | | | | |
Collapse
|
17
|
Sun JB, Mielcarek N, Lakew M, Grzych JM, Capron A, Holmgren J, Czerkinsky C. Intranasal Administration of a Schistosoma mansoni Glutathione S-Transferase-Cholera Toxoid Conjugate Vaccine Evokes Antiparasitic and Antipathological Immunity in Mice. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.2.1045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
Mucosal administration of Ags linked to cholera toxin B subunit (CTB) can induce both strong mucosal secretory IgA immune responses and peripheral T cell hyporeactivity. In this study, intranasal (i.n.) administration of CTB-conjugated Schistosoma mansoni 28-kDa GST (CTB-Sm28GST) was found to protect infected animals from schistosomiasis, especially from immunopathological complications associated with chronic inflammation. Worm burden and liver egg counts were reduced in infected animals treated with the CTB-Sm28GST conjugate as compared with mice infected only, or with mice treated with a control (CTB-OVA) conjugate. However, a more striking and consistent effect was that granuloma formations in liver and lungs of mice treated with CTB-Sm28GST were markedly suppressed. Such treatment was associated with reduced systemic delayed-type hypersensitivity and lymphocyte proliferative responses to Sm28GST. Production of IFN-γ, IL-3, and IL-5 by liver cells was also markedly reduced after i.n. treatment of CTB-Sm28GST, whereas IL-4 production was not impaired. Intranasal treatment of infected mice with CTB-Sm28GST increased IgG1-, IgG2a-, IgA-, and IgE-Ab-forming cell responses in liver in comparison with treatment with CTB-OVA, or free Sm28GST. Most importantly, mucosal treatment with CTB-Sm28GST significantly reduced animal mortality when administered to chronically infected mice. Our results suggest that it may be possible to design a therapeutic vaccine against schistosomiasis that both limits infection and suppresses parasite-induced pathology.
Collapse
Affiliation(s)
- Jia-Bin Sun
- *Department of Medical Microbiology and Immunology, University of Göteborg, Göteborg, Sweden
| | - Nathalie Mielcarek
- *Department of Medical Microbiology and Immunology, University of Göteborg, Göteborg, Sweden
| | - Mekuria Lakew
- *Department of Medical Microbiology and Immunology, University of Göteborg, Göteborg, Sweden
- ‡Department of Biology, Addis Ababa University, Addis Ababa, Ethiopia; and
| | - Jean-Marie Grzych
- †Centre d’Immunologie et de Biologie Parasitaire, Institut National de la Santé et de la Recherche Médicale U167, Institut Pasteur de Lille, Lille, France
| | - Andre Capron
- †Centre d’Immunologie et de Biologie Parasitaire, Institut National de la Santé et de la Recherche Médicale U167, Institut Pasteur de Lille, Lille, France
| | - Jan Holmgren
- *Department of Medical Microbiology and Immunology, University of Göteborg, Göteborg, Sweden
| | - Cecil Czerkinsky
- *Department of Medical Microbiology and Immunology, University of Göteborg, Göteborg, Sweden
- §Institut National de la Santé et de la Recherche Médicale U364, Nice, France
| |
Collapse
|
18
|
Riveau G, Poulain-Godefroy OP, Dupré L, Remoué F, Mielcarek N, Locht C, Capron A. Glutathione S-transferases of 28kDa as major vaccine candidates against schistosomiasis. Mem Inst Oswaldo Cruz 1999; 93 Suppl 1:87-94. [PMID: 9921328 DOI: 10.1590/s0074-02761998000700012] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
For the development of vaccine strategies to generate efficient protection against chronic infections such as parasitic diseases, and more precisely schistosomiasis, controlling pathology could be more relevant than controlling the infection itself. Such strategies, motivated by the need for a cost-effective complement to existing control measures, should focus on parasite molecules involved in fecundity, because in metazoan parasite infections pathology is usually linked to the output of viable eggs. In numerous animal models, vaccination with glutathione S-transferases of 28kDa has been shown to generate an immune response strongly limiting the worm fecundity, in addition to the reduction of the parasite burden. Recent data on acquired immunity directed to 28GST in infected human populations, and new development to draw adapted vaccine formulations, are presented.
Collapse
Affiliation(s)
- G Riveau
- Inserm U167, Institut Pasteur de Lille, France.
| | | | | | | | | | | | | |
Collapse
|
19
|
Angyalosi G, Pancré V, Herno J, Auriault C. Immunological response of major histocompatibility complex class II-deficient (Abeta(o)) mice infected by the parasite Schistosoma mansoni. Scand J Immunol 1998; 48:159-69. [PMID: 9716107 DOI: 10.1046/j.1365-3083.1998.00372.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We have characterized the immunological behaviour of major histocompitibility complex (MHC) Class II molecule-deficient (Abeta(o)) mice after infection by Schistosoma mansoni. In Abeta(o) mice, morbidity developed dramatically 7 weeks after infection leading to death, despite the absence of an increase in parasite burden or of eggs trapped in the liver. Histological examination of the liver revealed the absence of a classical granulomatous reaction. Antibodies were produced only against schistosomulum antigens. Specific antibodies against adult worm (SWAP) or egg antigen (SEA) were not detected. Cytokine production (IFN-gamma and IL-4) was absent after in vitro restimulation of splenic cells from infected Abeta(o) mice with parasite antigens. Adoptive transfer of primed splenic cells (total, purified CD4+ or CD8+ T cells) failed to improve survival or to induce a granulomatous reaction in infected Abeta(o) mice. Survival, cellular and humoral responses in CD8+ T-cell-depleted Abeta(o) mice or MHC(o) mice (lacking MHC class I and II molecules) were similar to nondepleted Abeta(o) mice, suggesting that anti-schistosomula antibody production was thymo-independent. Our results demonstrate a high degree of susceptibility of Abeta(o) mice to infection and corroborate the importance of CD4+ T cells in the initiation of the granulomatous response. However, our results do not show evidence for the involvement of CD8+ T cells in response to S. mansoni infection.
Collapse
Affiliation(s)
- G Angyalosi
- Laboratoire d'Immunologie Cellulaire de l'Interface Hôte/Parasite et de la Pathogénèse Parasitaire, URA-CNRS 1854, Institut Pasteur de Lille, France
| | | | | | | |
Collapse
|
20
|
|
21
|
Asseman C, Pancré V, Quatennens B, Auriault C. Schistosoma mansoni-infected mice show augmented hepatic fibrosis and selective inhibition of liver cytokine production after treatment with anti-NK1.1 antibodies. Immunol Lett 1996; 54:11-20. [PMID: 9030976 DOI: 10.1016/s0165-2478(96)02634-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Gamma interferon (IFN-gamma) plays an immunoregulatory role at different stages of the experimental Schistosoma mansoni-driven processes in mice through its ability to induce cell cytotoxicity against the parasite larvae and to reduce established hepatic fibrosis. The role of Natural Killer (NK) cells, as possible major source of IFN-gamma, has never been studied during the entire course of murine schistosomiasis. In this paper, we investigated the consequences of in vivo NK cell depletion, maintained during 17 weeks of infection, on both hepatic granuloma development and immunological parameters. We found that NK cell depletion following anti-NK1.1 monoclonal antibody (mAb) injections led to an increase of hepatic collagen content in the late stages of granuloma formation and to the diminution of interleukin 12 (IL-12) p40 and IL-7 mRNA expression in the livers. The hepatic mRNA expression of other cytokines (IFN-gamma, tumor necrosis factor alpha [TNF-alpha] and IL-4), as well as humoral and cytokine responses in sera, were not significantly different between control monoclonal antibody (CmAb) and anti-NK1.1-treated mice. Thus, we demonstrate that the anti-NK1.1 treatment might induce alterations of regulatory mechanisms, detectable at a late stage of a chronic process in immunocompetent mice.
Collapse
Affiliation(s)
- C Asseman
- URA CNRS 1854, Institut Pasteur, Lille, France
| | | | | | | |
Collapse
|
22
|
Pillai S, Dermody K, Metcalf B. Immunogenicity of genetically engineered glutathione S-transferase fusion proteins containing a T-cell epitope from diphtheria toxin. Infect Immun 1995; 63:1535-40. [PMID: 7534277 PMCID: PMC173186 DOI: 10.1128/iai.63.4.1535-1540.1995] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Glutathione S-transferase (GST) has been shown to induce a marginal antibody response in experimental animals as well as partial protection against a number of parasitic worms, including Schistosoma and Fasciola species. The objective of our study was to increase the immunogenicity of GST by adding heterologous T-cell epitopes at the carboxy terminus of the protein. We generated recombinant GST proteins by attaching one or three tandem repeats of a T-cell epitope of CRM197, a nontoxic variant of diphtheria toxin. This T-cell epitope encoding the region of amino acids 366 to 383 of CRM197, when contained in a GST fusion protein and/or after purification as a recombinant peptide, retained the ability to induce a CRM197-specific T-cell response. The fusion protein containing a single T-cell epitope induced a strong T-cell proliferative response to GST and also enhanced anti-GST antibody production in mice. The addition of three repeats of the epitope did not augment the responses when compared with the responses of GST itself. The results suggest that the addition of a single T-cell epitope to a larger protein like GST increases the immunogenicity of the protein.
Collapse
Affiliation(s)
- S Pillai
- Department of Immunology, Lederle-Praxis Biologicals Division, West Henrietta, New York 14586
| | | | | |
Collapse
|