1
|
Zumuk CP, Jones MK, Navarro S, Gray DJ, You H. Transmission-Blocking Vaccines against Schistosomiasis Japonica. Int J Mol Sci 2024; 25:1707. [PMID: 38338980 PMCID: PMC10855202 DOI: 10.3390/ijms25031707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
Control of schistosomiasis japonica, endemic in Asia, including the Philippines, China, and Indonesia, is extremely challenging. Schistosoma japonicum is a highly pathogenic helminth parasite, with disease arising predominantly from an immune reaction to entrapped parasite eggs in tissues. Females of this species can generate 1000-2200 eggs per day, which is about 3- to 15-fold greater than the egg output of other schistosome species. Bovines (water buffalo and cattle) are the predominant definitive hosts and are estimated to generate up to 90% of parasite eggs released into the environment in rural endemic areas where these hosts and humans are present. Here, we highlight the necessity of developing veterinary transmission-blocking vaccines for bovines to better control the disease and review potential vaccine candidates. We also point out that the approach to producing efficacious transmission-blocking animal-based vaccines before moving on to human vaccines is crucial. This will result in effective and feasible public health outcomes in agreement with the One Health concept to achieve optimum health for people, animals, and the environment. Indeed, incorporating a veterinary-based transmission vaccine, coupled with interventions such as human mass drug administration, improved sanitation and hygiene, health education, and snail control, would be invaluable to eliminating zoonotic schistosomiasis.
Collapse
Affiliation(s)
- Chika P. Zumuk
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (C.P.Z.); (M.K.J.); (S.N.)
- Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
| | - Malcolm K. Jones
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (C.P.Z.); (M.K.J.); (S.N.)
- School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia
| | - Severine Navarro
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (C.P.Z.); (M.K.J.); (S.N.)
- Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
- Centre for Childhood Nutrition Research, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Darren J. Gray
- Population Health Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia;
| | - Hong You
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; (C.P.Z.); (M.K.J.); (S.N.)
- School of Veterinary Science, The University of Queensland, Gatton, QLD 4343, Australia
| |
Collapse
|
2
|
Muslimov A, Tereshchenko V, Shevyrev D, Rogova A, Lepik K, Reshetnikov V, Ivanov R. The Dual Role of the Innate Immune System in the Effectiveness of mRNA Therapeutics. Int J Mol Sci 2023; 24:14820. [PMID: 37834268 PMCID: PMC10573212 DOI: 10.3390/ijms241914820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/24/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Advances in molecular biology have revolutionized the use of messenger RNA (mRNA) as a therapeutic. The concept of nucleic acid therapy with mRNA originated in 1990 when Wolff et al. reported successful expression of proteins in target organs by direct injection of either plasmid DNA or mRNA. It took decades to bring the transfection efficiency of mRNA closer to that of DNA. The next few decades were dedicated to turning in vitro-transcribed (IVT) mRNA from a promising delivery tool for gene therapy into a full-blown therapeutic modality, which changed the biotech market rapidly. Hundreds of clinical trials are currently underway using mRNA for prophylaxis and therapy of infectious diseases and cancers, in regenerative medicine, and genome editing. The potential of IVT mRNA to induce an innate immune response favors its use for vaccination and immunotherapy. Nonetheless, in non-immunotherapy applications, the intrinsic immunostimulatory activity of mRNA directly hinders the desired therapeutic effect since it can seriously impair the target protein expression. Targeting the same innate immune factors can increase the effectiveness of mRNA therapeutics for some indications and decrease it for others, and vice versa. The review aims to present the innate immunity-related 'barriers' or 'springboards' that may affect the development of immunotherapies and non-immunotherapy applications of mRNA medicines.
Collapse
Affiliation(s)
- Albert Muslimov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
- Laboratory of Nano- and Microencapsulation of Biologically Active Substances, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia;
- RM Gorbacheva Research Institute, Pavlov University, L’va Tolstogo 6-8, 197022 St. Petersburg, Russia;
| | - Valeriy Tereshchenko
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| | - Daniil Shevyrev
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| | - Anna Rogova
- Laboratory of Nano- and Microencapsulation of Biologically Active Substances, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, 195251 St. Petersburg, Russia;
- Saint-Petersburg Chemical-Pharmaceutical University, Professora Popova 14, 197376 St. Petersburg, Russia
- School of Physics and Engineering, ITMO University, Lomonosova 9, 191002 St. Petersburg, Russia
| | - Kirill Lepik
- RM Gorbacheva Research Institute, Pavlov University, L’va Tolstogo 6-8, 197022 St. Petersburg, Russia;
| | - Vasiliy Reshetnikov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Prospekt Akad. Lavrentyeva 10, 630090 Novosibirsk, Russia
| | - Roman Ivanov
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, Olympic Ave 1, 354340 Sirius, Russia; (V.T.); (D.S.); (V.R.); (R.I.)
| |
Collapse
|
3
|
Schistosoma mansoni Adult Worm Protective and Diagnostic Proteins in n-Butanol Extracts Revealed by Proteomic Analysis. Pathogens 2021; 11:pathogens11010022. [PMID: 35055970 PMCID: PMC8777762 DOI: 10.3390/pathogens11010022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/09/2021] [Accepted: 12/21/2021] [Indexed: 12/02/2022] Open
Abstract
The S. mansoni adult worm n-butanol extract (Sm-AWBE) has been previously shown to contain specific S. mansoni antigens that have been used for immunodiagnosis of schistosomiasis in solid phase alkaline phosphatase immunoassay (APIA) and western blot (WB) analyses. Sm-AWBE was also used in immunoprotection studies against a fatal live-cercariae challenge in experimental mouse vaccination (~43% protection). The Sm-AWBE fraction was prepared by mixing adult worm membranous suspensions with aqueous-saturated n-butanol, centrifuging and recovering n-butanol-resistant proteins in the aqueous phase. Here we report a preliminary identification of Sm-AWBE protein components as revealed from a qualitative proteomic study after processing Sm-AWBE by 1D-gel electrophoresis, in-gel and in-solution tryptic digestions, and mass spectrometry analyses. We identified 33 proteins in Sm-AWBE, all previously known S. mansoni proteins and antigens; among them, immunomodulatory proteins and proteins mostly involved in host–parasite interactions. About 81.8% of the identified Sm-AWBE proteins are antigenic. STRING analysis showed a set of Sm-AWBE proteins configuring a small network of interactive proteins and a group of proteins without interactions. Functional groups of proteins included muscle contraction, antioxidant, GPI-anchored phosphoesterases, regulatory 14-3-3, various enzymes and stress proteins. The results widen the possibilities to design novel antigen combinations for better diagnostic and immunoprotective strategies for schistosomiasis control.
Collapse
|
4
|
Immunization of Goats with Recombinant Protein 14-3-3 Isoform 2(rHcftt-2) Induced Moderate Protection against Haemonchus contortus Challenge. Pathogens 2020; 9:pathogens9010046. [PMID: 31935869 PMCID: PMC7168593 DOI: 10.3390/pathogens9010046] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/05/2020] [Accepted: 01/05/2020] [Indexed: 12/13/2022] Open
Abstract
A previous study identified that isoform 2 (Hcftt-2) of the 14-3-3 protein of Haemonchus contortus (H. contortus) could suppress immune functions of goat peripheral blood mononuclear cells (PBMCs) and might be a potential vaccine target, as neutralization of the protein function may enhance anti-parasite immunity. In this research, the recombinant Hcftt-2 was evaluated for its immunoprotective efficacy against H. contortus infection in goats. Five experimental goats were immunized twice with rHcftt-2 along with Freund’s adjuvant. The five immunized goats and five nonimmunized goats (adjuvant only) were challenged with 5000 L3-stage H. contortus larvae after 14 days of second immunization. Five nonimmunized and uninfected goats (adjuvant only) were set as the uninfected group. A significant increase in the serum immunoglobin G(IgG) and serum IgA levels were identified in the rHcftt-2 immunized animals. The mean eggs per gram in feces (EPG) and the worm burdens of rHcftt-2 immunized group were reduced by 26.46% (p < 0.05) and 32.33%, respectively. In brief, immunization of goats with rHcftt-2 induced moderate protection against H. contortus challenge.
Collapse
|
5
|
Masoori L, Falak R, Mokhtarian K, Bandehpour M, Razmjou E, Jalallou N, Jafarian F, Akhlaghi L, Meamar AR. Production of recombinant 14-3-3 protein and determination of its immunogenicity for application in serodiagnosis of strongyloidiasis. Trans R Soc Trop Med Hyg 2019; 113:326-331. [DOI: 10.1093/trstmh/trz006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/16/2018] [Accepted: 02/03/2019] [Indexed: 12/14/2022] Open
Affiliation(s)
- Leila Masoori
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Kobra Mokhtarian
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mojgan Bandehpour
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Razmjou
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nahid Jalallou
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Farzane Jafarian
- Ayatollah Rouhani Hospital, Babol University of Medical Sciences, Mazandaran, Iran
| | - Lame Akhlaghi
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Meamar
- Department of Parasitology and Mycology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Młocicki D, Sulima A, Bień J, Näreaho A, Zawistowska-Deniziak A, Basałaj K, Sałamatin R, Conn DB, Savijoki K. Immunoproteomics and Surfaceomics of the Adult Tapeworm Hymenolepis diminuta. Front Immunol 2018; 9:2487. [PMID: 30483248 PMCID: PMC6240649 DOI: 10.3389/fimmu.2018.02487] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 10/08/2018] [Indexed: 12/20/2022] Open
Abstract
In cestodiasis, mechanical and molecular contact between the parasite and the host activates the immune response of the host and may result in inflammatory processes, leading to ulceration and intestinal dysfunctions. The aim of the present study was to identify antigenic proteins of the adult cestode Hymenolepis diminuta by subjecting the total protein extracts from adult tapeworms to 2DE immunoblotting (two-dimensional electrophoresis combined with immunoblotting) using sera collected from experimentally infected rats. A total of 36 protein spots cross-reacting with the rat sera were identified using LC-MS/MS. As a result, 68 proteins, including certain structural muscle proteins (actin, myosin, and paramyosin) and moonlighters (heat shock proteins, kinases, phosphatases, and glycolytic enzymes) were identified; most of these were predicted to possess binding and/or catalytic activity required in various metabolic and cellular processes, and reported here as potential antigens of the adult cestode for the first time. As several of these antigens can also be found at the cell surface, the surface-associated proteins were extracted and subjected to in-solution digestion for LC-MS/MS identification (surfaceomics). As a result, a total of 76 proteins were identified, from which 31 proteins, based on 2DE immunoblotting, were predicted to be immunogenic. These included structural proteins actin, myosin and tubulin as well as certain moonlighting proteins (heat-shock chaperones) while enzymes with diverse catalytic activities were found as the most dominating group of proteins. In conclusion, the present study shed new light into the complexity of the enteric cestodiasis by showing that the H. diminuta somatic proteins exposed to the host possess immunomodulatory functions, and that the immune response of the host could be stimulated by diverse mechanisms, involving also those triggering protein export via yet unknown pathways.
Collapse
Affiliation(s)
- Daniel Młocicki
- Department of General Biology and ParasitologyMedical University of Warsaw, Warsaw, Poland
- Witold Stefański Institute of ParasitologyPolish Academy of Sciences, Warsaw, Poland
| | - Anna Sulima
- Department of General Biology and ParasitologyMedical University of Warsaw, Warsaw, Poland
| | - Justyna Bień
- Witold Stefański Institute of ParasitologyPolish Academy of Sciences, Warsaw, Poland
| | - Anu Näreaho
- Department of Veterinary BiosciencesUniversity of Helsinki, Helsinki, Finland
| | | | - Katarzyna Basałaj
- Witold Stefański Institute of ParasitologyPolish Academy of Sciences, Warsaw, Poland
| | - Rusłan Sałamatin
- Department of General Biology and ParasitologyMedical University of Warsaw, Warsaw, Poland
- Department of Parasitology and Vector-Borne DiseasesNational Institute of Public Health–National Institute of Hygiene, Warsaw, Poland
| | - David Bruce Conn
- Department of Invertebrate Zoology, Museum of Comparative Zoology, Harvard UniversityCambridge, MA, United States
- One Health Center, Berry CollegeMount Berry, GA, United States
| | - Kirsi Savijoki
- Division of Pharmaceutical BiosciencesUniversity of Helsinki, Helsinki, Finland
| |
Collapse
|
7
|
Flagellin-deficient outer membrane vesicles as adjuvant induce cross-protection of Salmonella Typhimurium outer membrane proteins against infection by heterologous Salmonella serotypes. Int J Med Microbiol 2018; 308:796-802. [DOI: 10.1016/j.ijmm.2018.06.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/30/2018] [Accepted: 06/03/2018] [Indexed: 12/23/2022] Open
|
8
|
Pathological, immunological and parasitological study of sheep vaccinated with the recombinant protein 14-3-3z and experimentally infected with Fasciola hepatica. Vet Immunol Immunopathol 2018; 202:115-121. [PMID: 30078585 DOI: 10.1016/j.vetimm.2018.07.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/17/2018] [Accepted: 07/07/2018] [Indexed: 11/23/2022]
Abstract
In this study, the immunogenicity and protective capacity of a new recombinant vaccine candidate, the rFh14-3-3z protein was analysed in sheep experimentally challenged with Fasciola hepatica, in terms of fluke burden, faecal egg counts, hepatic damage and humoral immune response. Three groups of 8 animals each were used for study, group 1 was immunised with the rFh14-3-3z in Montanide adjuvant, whereas group 2 and 3 remained as adjuvant control and infection control groups, respectively. The parasitological analysis showed that no significant reduction in fluke burden, fluke size and faecal egg counts was detected. The extent of hepatic damage was very similar between groups. Nonetheless, animals immunised with the rFh14-3-3z protein induced the development of specific IgG1 and IgG2, being the IgG1 the predominant antibody; which confirms the immunogenicity of this protein in sheep. This is the first report of the 14-3-3z proteins as vaccine against the infection with F. hepatica.
Collapse
|
9
|
Sulima A, Bień J, Savijoki K, Näreaho A, Sałamatin R, Conn DB, Młocicki D. Identification of immunogenic proteins of the cysticercoid of Hymenolepis diminuta. Parasit Vectors 2017; 10:577. [PMID: 29157281 PMCID: PMC5697066 DOI: 10.1186/s13071-017-2519-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 11/01/2017] [Indexed: 01/03/2023] Open
Abstract
Background A wide range of molecules are used by tapeworm metacestodes to establish successful infection in the hostile environment of the host. Reports indicating the proteins in the cestode-host interactions are limited predominantly to taeniids, with no previous data available for non-taeniid species. A non-taeniid, Hymenolepis diminuta, represents one of the most important model species in cestode biology and exhibits an exceptional developmental plasticity in its life-cycle, which involves two phylogenetically distant hosts, arthropod and vertebrate. Results We identified H. diminuta cysticercoid proteins that were recognized by sera of H. diminuta-infected rats using two-dimensional gel electrophoresis (2DE), 2D-immunoblotting, and LC-MS/MS mass spectrometry. Proteomic analysis of 42 antigenic spots revealed 70 proteins. The largest number belonged to structural proteins and to the heat-shock protein (HSP) family. These results show a number of the antigenic proteins of the cysticercoid stage, which were present already in the insect host prior to contact with the mammal host. These are the first parasite antigens that the mammal host encounters after the infection, therefore they may represent some of the molecules important in host-parasite interactions at the early stage of infection. Conclusions These results could help in understanding how H. diminuta and other cestodes adapt to their diverse and complex parasitic life-cycles and show universal molecules used among diverse groups of cestodes to escape the host response to infection. Electronic supplementary material The online version of this article (10.1186/s13071-017-2519-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anna Sulima
- Department of General Biology and Parasitology, Medical University of Warsaw, Warsaw, Poland
| | - Justyna Bień
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Warsaw, Poland
| | - Kirsi Savijoki
- Department of Food and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Anu Näreaho
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Rusłan Sałamatin
- Department of General Biology and Parasitology, Medical University of Warsaw, Warsaw, Poland.,Department of Medical Parasitology, National Institute of Public Health - National Institute of Hygiene, Warsaw, Poland
| | - David Bruce Conn
- One Health Center, Berry College, Mount Berry, GA, USA.,Department of Invertebrate Zoology, Museum of Comparative Zoology, Harvard University, Cambridge, MA, USA
| | - Daniel Młocicki
- Department of General Biology and Parasitology, Medical University of Warsaw, Warsaw, Poland. .,Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
10
|
Yang J, Zhu W, Huang J, Wang X, Sun X, Zhan B, Zhu X. Partially protective immunity induced by the 14-3-3 protein from Trichinella spiralis. Vet Parasitol 2016; 231:63-68. [PMID: 27387374 DOI: 10.1016/j.vetpar.2016.06.028] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 06/12/2016] [Accepted: 06/22/2016] [Indexed: 12/22/2022]
Abstract
Trichinellosis, a widespread zoonosis, is considered to be an emerging or re-emerging infectious parasitic disease. The development of vaccines to prevent Trichinella infection in domestic animals and humans is important for disease control. In a previous study, we identified Ts14-3-3 as an immunodominant protein from Trichinella spiralis (T. spiralis) adult worms recognized by early infection sera from pigs and mice. In this study, we further confirmed that Ts14-3-3 mRNA is expressed in both adult worms and in the larval stages of T. spiralis. Immunostaining with anti-Ts14-3-3 mouse sera further confirmed that native Ts14-3-3 is highly expressed on the surface of T. spiralis muscle larvae. The immune recognition by infected sera, its expression in both adult and larval stages and its exposure on the surface of the parasite led us to explore Ts14-3-3 as a vaccine antigen. Recombinant Ts14-3-3 formulated with an ISA50v2 adjuvant produced strong total IgG and balanced IgG1 and IgG2a responses in vaccinated mice and stimulated mouse splenocytes to produce high levels of Th1 (INF-γ, IL-2) and Th2 (IL4, IL5) cytokines. These results indicate that Ts14-3-3 is highly immunogenic and is able to induce balanced Th1/Th2 immune responses. These vaccine-induced immune responses resulted in a reduction in muscle larvae of up to 46.2% in vaccinated mice upon subsequent larval challenge relative to the number of larvae in mice received PBS control. The significant reduction in muscle larvae in vaccinated mice suggests that Ts14-3-3 is a promising vaccine target for potential use in domestic pigs to prevent trichinellosis transmission.
Collapse
Affiliation(s)
- Jing Yang
- Department of Medical Microbiology & Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| | - Wei Zhu
- Department of Medical Microbiology & Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| | - Jingjing Huang
- Department of Medical Microbiology & Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| | - Xiaohuan Wang
- Department of Medical Microbiology & Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| | - Ximeng Sun
- Department of Medical Microbiology & Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| | - Bin Zhan
- Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, 9 Houston, TX 77030, USA.
| | - Xinping Zhu
- Department of Medical Microbiology & Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| |
Collapse
|
11
|
Cheng PC, Lin CN, Peng SY, Kang TF, Lee KM. Combined IL-12 Plasmid and Recombinant SjGST Enhance the Protective and Anti-pathology Effect of SjGST DNA Vaccine Against Schistosoma japonicum. PLoS Negl Trop Dis 2016; 10:e0004459. [PMID: 26891172 PMCID: PMC4758724 DOI: 10.1371/journal.pntd.0004459] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/23/2016] [Indexed: 01/10/2023] Open
Abstract
Schistosomiasis is listed as one of most important tropical diseases and more than 200 million people are estimated to be infected. Development of a vaccine is thought to be the most effective way to control this disease. Recombinant 26-kDa glutathione S-transferase (rSjGST) has previously been reported to achieve a worm reduction rate of 42-44%. To improve the efficiency of the vaccine against Schistosoma japonicum, we immunized mice with a combination of pcDNA vector-encoded 26-kDa SjGST (pcDNA/SjGST), IL-12 expressing-plasmid (pIL-12), and rSjGST. Co-vaccination with pcDNA/SjGST, pIL-12, and rSjGST led to a reduction in worm burden, hepatic egg burden, and the size of liver tissue granulomas than that in the untreated infection controls. In addition, we detected high levels of specific IgG, IgG1, and IgG2a against the rSjGST antigen in infected mice vaccinated with this combination of pcDNA/SjGST, pIL-12, and rSjGST. Moreover, high expression levels of Th2 cytokines, including IL-4 and IL-10, were also detected in this group, without diminished levels of IL-12, INF-γ, and TNF-α cytokines that are related to parasite killing. In conclusion, we have developed a new vaccination regimen against S. japonicum infection and shown that co-immunization with pcDNA/SjGST vaccine, pIL-12, and rSjGST has significant anti-parasite, anti-hepatic egg and anti-pathology effects in mice. The efficacy of this vaccination method should be further validated in large animals such as water buffalo. This method may help to reduce the transmission of zoonotic schistosomiasis japonica.
Collapse
Affiliation(s)
- Po-Ching Cheng
- Department of Molecular Parasitology and Tropical Diseases, School of Medicine, Taipei Medical University, Taipei, Taiwan
- * E-mail: (PCC); (KML)
| | - Ching-Nan Lin
- Institute of Microbiology and Immunology, National Yang-Mng University, Taipei, Taiwan
- Institute of Tropical Medicine, National Yang-Mng University, Taipei, Taiwan
| | - Shih-Yi Peng
- Department of Biochemistry, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Tsung-Fu Kang
- Institute of Tropical Medicine, National Yang-Mng University, Taipei, Taiwan
| | - Kin-Mu Lee
- Institute of Biotechnology, Central Taiwan University of Science and Technology, Taichung, Taiwan
- * E-mail: (PCC); (KML)
| |
Collapse
|
12
|
Mbanefo EC, Kumagai T, Kodama Y, Kurosaki T, Furushima-Shimogawara R, Cherif MS, Mizukami S, Kikuchi M, Huy NT, Ohta N, Sasaki H, Hirayama K. Immunogenicity and anti-fecundity effect of nanoparticle coated glutathione S-transferase (SjGST) DNA vaccine against murine Schistosoma japonicum infection. Parasitol Int 2015; 64:24-31. [PMID: 25603531 DOI: 10.1016/j.parint.2015.01.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 12/10/2014] [Accepted: 01/11/2015] [Indexed: 01/10/2023]
Abstract
There is still urgent need for a vaccine against schistosomiasis, especially in Schistosoma japonicum endemic areas where even a vaccine that will interrupt zoonotic transmission will be potentially effective as an intervention tool. We had developed a novel nanoparticle gene delivery system, which has proven efficacious in gene transfection to target immune cells with complementary adjuvant effect and high protective efficacy in several diseases. Here, we applied this nanoparticle system in combination with S. japonicum glutathione S-transferase (SjGST) DNA vaccine to show the immunogenicity and anti-fecundity effect of the nanoparticle coated vaccine formulation against murine schistosomiasis. The nanoparticle-coated DNA vaccine formulation induced desired immune responses. In comparison with the nanoparticle coated empty vector, it produced significantly increased antigen-specific humoral response, T-helper 1 polarized cytokine environment, higher proportion of IFN-γ producing CD4(+) T-cells and the concomitant decrease in IL-4 producing CD4(+) T-cells. Although there was no effect on worm burden, we recorded a marked reduction in tissue egg burden. There was up to 71.3% decrease in tissue egg burden and 55% reduction in the fecundity of female adult worms. Our data showed that SjGST DNA vaccine, delivered using the nanoparticle gene delivery system, produced anti-fecundity effect on female adult schistosomes as previously described by using conventional subunit vaccine with adjuvant, proving this DNA vaccine formulation as a promising candidate for anti-pathology and transmission blocking application.
Collapse
Affiliation(s)
- Evaristus Chibunna Mbanefo
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN) and Global COE Program, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Department of Parasitology and Entomology, Faculty of Bioscience, Nnamdi Azikiwe University, P.M.B. 5025, Awka, Nigeria
| | - Takashi Kumagai
- Section of Environmental Parasitology, Tokyo Medical and Dental University Graduate School of Medical and Dental Science, 113-8519, Japan
| | - Yukinobu Kodama
- Department of Hospital Pharmacy, Nagasaki University Hospital, 1-7-1 Sakamoto, 852-8501, Japan
| | - Tomoaki Kurosaki
- Department of Hospital Pharmacy, Nagasaki University Hospital, 1-7-1 Sakamoto, 852-8501, Japan
| | - Rieko Furushima-Shimogawara
- Section of Environmental Parasitology, Tokyo Medical and Dental University Graduate School of Medical and Dental Science, 113-8519, Japan
| | - Mahamoud Sama Cherif
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN) and Global COE Program, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan
| | - Shusaku Mizukami
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN) and Global COE Program, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan
| | - Mihoko Kikuchi
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN) and Global COE Program, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan
| | - Nguyen Tien Huy
- Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Department of Clinical Product Development, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan
| | - Nobuo Ohta
- Section of Environmental Parasitology, Tokyo Medical and Dental University Graduate School of Medical and Dental Science, 113-8519, Japan
| | - Hitoshi Sasaki
- Department of Hospital Pharmacy, Nagasaki University Hospital, 1-7-1 Sakamoto, 852-8501, Japan
| | - Kenji Hirayama
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN) and Global COE Program, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan; Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, 852-8523, Japan.
| |
Collapse
|
13
|
Mbanefo EC, Huy NT, Wadagni AA, Eneanya CI, Nwaorgu O, Hirayama K. Host determinants of reinfection with schistosomes in humans: a systematic review and meta-analysis. PLoS Negl Trop Dis 2014; 8:e3164. [PMID: 25211227 PMCID: PMC4161334 DOI: 10.1371/journal.pntd.0003164] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Accepted: 08/04/2014] [Indexed: 12/12/2022] Open
Abstract
Background Schistosomiasis is still a major public health burden in the tropics and subtropics. Although there is an effective chemotherapy (Praziquantel) for this disease, reinfection occurs rapidly after mass drug administration (MDA). Because the entire population do not get reinfected at the same rate, it is possible that host factors may play a dominant role in determining resistance or susceptibility to reinfection with schistosomes. Here, we systematically reviewed and meta-analyzed studies that reported associations between reinfection with the principal human-infecting species (S. mansoni, S. japonicum and S. haematobium) and host socio-demographic, epidemiological, immunological and genetic factors. Methodology/Principal Findings PubMed, Scopus, Google Scholar, Cochrane Review Library and African Journals Online public databases were searched in October 2013 to retrieve studies assessing association of host factors with reinfection with schistosomes. Meta-analysis was performed to generate pooled odds ratios and standardized mean differences as overall effect estimates for dichotomous and continuous variables, respectively. Quality assessment of included studies, heterogeneity between studies and publication bias were also assessed. Out of the initial 2739 records, 109 studies were included in the analyses, of which only 32 studies with 37 data sets were eligible for quantitative data synthesis. Among several host factors identified, strong positive association was found with age and pre-treatment intensity, and only slightly for gender. These factors are major determinants of exposure and disease transmission. Significant positive association was found with anti-SWA IgG4 level, and a negative overall effect for association with IgE levels. This reconfirmed the concept that IgE/IgG4 balance is a major determinant of protective immunity against schistosomiasis. Other identified determinants were reported by a small number of studies to enable interpretation. Conclusions Our data contribute to the understanding of host-parasite interaction as it affects reinfection, and is a potential tool to guide planning and tailoring of community interventions to target high-risk groups. One of the major challenges of schistosomiasis control is that disease prevalence reverts to baseline levels after mass drug administration due to high rate of reinfection. Host factors play a major role in determining resistance or susceptibility to reinfection with schistosomiasis and other diseases. We systematically searched and analyzed studies that identified potential host determinants of reinfection with schistosomes. Among demographic variables, age but not gender was strongly associated with reinfection with schistosomes. Pretreatment infection intensity was also identified as a major determinant of reinfection. Positive association with IgG4 levels and negative association with IgE levels reconfirmed the notion that IgE/IgG4 balance is the major factor controlling protective immunity against schistosomiasis. Other factors were reported by few studies to allow correct inferences. These results contribute to our understanding of host-parasite relationship as it affects reinfection, and will be useful for planning and targeting the limited resources for intervention on high-risk groups.
Collapse
Affiliation(s)
- Evaristus Chibunna Mbanefo
- Department of Parasitology and Entomology, Faculty of Bioscience, Nnamdi Azikiwe University, Awka, Nigeria
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
- * E-mail: ,
| | - Nguyen Tien Huy
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
| | - Anita Akpeedje Wadagni
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
| | - Christine Ifeoma Eneanya
- Department of Parasitology and Entomology, Faculty of Bioscience, Nnamdi Azikiwe University, Awka, Nigeria
| | - Obioma Nwaorgu
- Department of Parasitology and Entomology, Faculty of Bioscience, Nnamdi Azikiwe University, Awka, Nigeria
| | - Kenji Hirayama
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
| |
Collapse
|
14
|
Stephenson R, You H, McManus DP, Toth I. Schistosome Vaccine Adjuvants in Preclinical and Clinical Research. Vaccines (Basel) 2014; 2:654-85. [PMID: 26344751 PMCID: PMC4494218 DOI: 10.3390/vaccines2030654] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 08/08/2014] [Accepted: 08/15/2014] [Indexed: 12/16/2022] Open
Abstract
There is currently no vaccine available for human use for any parasitic infections, including the helminth disease, schistosomiasis. Despite many researchers working towards this goal, one of the focuses has been on identifying new antigenic targets. The bar to achieve protective efficacy in humans was set at a consistent induction of 40% protection or better by the World Health Organisation (WHO), and although this is a modest goal, it is yet to be reached with the six most promising schistosomiasis vaccine candidates (Sm28GST, IrV5, Sm14, paramyosin, TPI, and Sm23). Adjuvant selection has a large impact on the effectiveness of the vaccine, and the use of adjuvants to aid in the stimulation of the immune system is a critical step and a major variable affecting vaccine development. In addition to a comprehensive understanding of the immune system, level of protection and the desired immune response required, there is also a need for a standardised and effective adjuvant formulation. This review summarises the status of adjuvants that have been or are being employed in schistosomiasis vaccine development focusing on immunisation outcomes at preclinical and clinical stages.
Collapse
Affiliation(s)
- Rachel Stephenson
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia.
| | - Hong You
- Molecular Parasitology Laboratory, Infectious Diseases Division, QIMR Berghofer Medical Research Institute, Brisbane, Queensland Q4006, Australia.
| | - Donald P McManus
- Molecular Parasitology Laboratory, Infectious Diseases Division, QIMR Berghofer Medical Research Institute, Brisbane, Queensland Q4006, Australia.
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia.
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia.
| |
Collapse
|
15
|
Dias SRC, Boroni M, Rocha EA, Dias TL, de Laet Souza D, Oliveira FMS, Bitar M, Macedo AM, Machado CR, Caliari MV, Franco GR. Evaluation of the Schistosoma mansoni Y-box-binding protein (SMYB1) potential as a vaccine candidate against schistosomiasis. Front Genet 2014; 5:174. [PMID: 24966869 PMCID: PMC4052899 DOI: 10.3389/fgene.2014.00174] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 05/22/2014] [Indexed: 12/21/2022] Open
Abstract
Schistosomiasis is a neglected tropical disease, and after malaria, is the second most important tropical disease in public health. A vaccine that reduces parasitemia is desirable to achieve mass treatment with a low cost. Although potential antigens have been identified and tested in clinical trials, no effective vaccine against schistosomiasis is available. Y-box-binding proteins (YBPs) regulate gene expression and participate in a variety of cellular processes, including transcriptional and translational regulation, DNA repair, cellular proliferation, drug resistance, and stress responses. The Schistosoma mansoni ortholog of the human YB-1, SMYB1, is expressed in all stages of the parasite life cycle. Although SMYB1 binds to DNA or RNA oligonucleotides, immunohistochemistry assays demonstrated that it is primarily localized in the cytoplasm of parasite cells. In addition, SMYB1 interacts with a protein involved in mRNA processing, suggesting that SMYB1 functions in the turnover, transport, and/or stabilization of RNA molecules during post-transcriptional gene regulation. Here we report the potential of SMYB1 as a vaccine candidate. We demonstrate that recombinant SMYB1 stimulates the production of high levels of specific IgG1 antibodies in a mouse model. The observed levels of specific IgG1 and IgG2a antibodies indicate an actual protection against cercariae challenge. Animals immunized with rSMYB1 exhibited a 26% reduction in adult worm burden and a 28% reduction in eggs retained in the liver. Although proteins from the worm tegument are considered optimal targets for vaccine development, this study demonstrates that unexposed cytoplasmic proteins can reduce the load of intestinal worms and the number of eggs retained in the liver.
Collapse
Affiliation(s)
- Sílvia R C Dias
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte, Brazil
| | - Mariana Boroni
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte, Brazil
| | - Elizângela A Rocha
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte, Brazil
| | - Thomaz L Dias
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte, Brazil
| | - Daniela de Laet Souza
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte, Brazil
| | - Fabrício M S Oliveira
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte, Brazil
| | - Mainá Bitar
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte, Brazil
| | - Andrea M Macedo
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte, Brazil
| | - Carlos R Machado
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte, Brazil
| | - Marcelo V Caliari
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte, Brazil
| | - Glória R Franco
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte, Brazil
| |
Collapse
|
16
|
Tu Y, Hu Y, Fan G, Chen Z, Liu L, Man D, Liu S, Tang C, Zhang Y, Dai W. Protective effects of membrane-anchored and secreted DNA vaccines encoding fatty acid-binding protein and glutathione S-transferase against Schistosoma japonicum. PLoS One 2014; 9:e86575. [PMID: 24466157 PMCID: PMC3900569 DOI: 10.1371/journal.pone.0086575] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 12/12/2013] [Indexed: 01/09/2023] Open
Abstract
In order to explore the high performance bivalent DNA-based vaccine against schistosomes, SjFABP and Sj26GST were selected and used to construct a vaccine. Two strategies were used to construct the bivalent DNA vaccine. In the first strategy, a plasmid encoding antigen in the secreted form was used, while in the other, a plasmid encoding a truncated form of SjFABP and Sj26GST targeted to the cell surface was used. Various parameters, including antibody and cytokine response, proliferation, histopathological examination, and characterization of T cell subsets were used to evaluate the type of immune response and the level of protection against challenge infection. Injection with secreted pIRES-sjFABP-sj26GST significantly increased the levels of antibody, splenocyte proliferation, and production of IFN-γ, compared with membrane-anchored groups. Analysis of splenic T cell subsets showed that the secreted vaccine significantly increased the percentage of CD3+CD4+ and CD3+CD8+ T cells. Liver immunopathology (size of liver granulomas) was significantly reduced in the secreted group compared with the membrane-anchored groups. Moreover, challenge experiments showed that the worm and egg burdens were significantly reduced in animals immunized with recombinant vaccines. Most importantly, secreted Sj26GST-SjFABP markedly enhanced protection, by reducing worm and egg burdens by 31.8% and 24.78%, respectively, while the membrane-anchored group decreased worm and egg burdens by 24.80% and 18.80%, respectively. Taken together, these findings suggest that the secretory vaccine is more promising than the membrane-anchored vaccine, and provides support for the development and application of this vaccine.
Collapse
Affiliation(s)
- Yaqin Tu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yang Hu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- * E-mail: (WD); (YH)
| | - Guorun Fan
- Department of Otorhinolaryngology, Union Hospital of Tongji Medical College, Huazhong Science and Technology University, Wuhan, Hubei, China
| | - Zhihao Chen
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lin Liu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dandan Man
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuojie Liu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chengwu Tang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yin Zhang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wuxing Dai
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- * E-mail: (WD); (YH)
| |
Collapse
|
17
|
Buonaguro FM, Tornesello ML, Buonaguro L. New adjuvants in evolving vaccine strategies. Expert Opin Biol Ther 2011; 11:827-32. [PMID: 21609186 DOI: 10.1517/14712598.2011.587802] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adjuvants are becoming the key players of vaccine formulations to enhance the immunogenicity of subunit (peptides, proteins, virus-like particles (VLPs)) and DNA vaccines, as well as to reach the current new goals of preventing and/or treating chronic infectious diseases and cancers. Induction of humoral response, in particular neutralizing antibodies able to inhibit the binding of pathogens to their cellular receptors, remains a major goal of vaccines targeted to prevent acute lytic infections; induction/modulation of cellular immunity is, however, critical to fight latently/chronically infected cells as well as cancer cells. The new adjuvants, included in vaccine preparations, are currently able to modify the presentation of epitopes to the immune system with a specific T(H)1 versus T(H)2 polarization efficacy. A paradigm of the relevance of these new adjuvants is the immunological result obtained with the inclusion of monophosphoryl lipid A in the formulation of L1-based human papillomavirus (HPV)-naked VLPs. In the May issue of this journal, Garcon and colleagues describe the highly enhanced humoral and memory B cellular immunity of the AS04-adjuvanted HPV vaccine, which results in a long-lasting and broad spectrum immunity.
Collapse
Affiliation(s)
- Franco M Buonaguro
- Molecular Biology and Viral Oncology, Dpt of Experimental Oncology, Istituto Nazionale Tumori Fond Pascale, Napoli, Italy.
| | | | | |
Collapse
|
18
|
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
|
19
|
Wei F, Zhai Y, Jin H, Shang L, Men J, Lin J, Fu Z, Shi Y, Zhu XQ, Liu Q, Gao H. Development and immunogenicity of a recombinant pseudorabies virus expressing Sj26GST and SjFABP from Schistosoma japonicum. Vaccine 2010; 28:5161-6. [PMID: 20561603 DOI: 10.1016/j.vaccine.2010.06.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2010] [Revised: 06/02/2010] [Accepted: 06/03/2010] [Indexed: 11/27/2022]
Abstract
Recombinant pseudorabies virus (PRV) Bartha-K61 vaccine strains expressing Schistosoma japonicum 26kDa glutathione S-transferase (Sj26GST) and fatty acid binding protein (SjFABP), designated as rPRV/Sj26GST, rPRV/SjFABP and rPRV/Sj26GST-SjFABP, were constructed and evaluated for their ability to protect mice and sheep against S. japonicum challenge. Animals were given 2 intramuscular immunizations 3 weeks apart, and challenged with S. japonicum cercariae 4 weeks later. All mice vaccinated with recombinant virus developed specific anti-SWAP (soluble worm antigen preparation) antibody, splenocyte proliferative response and production of IFN-gamma and IL-2. Injection of rPRV/Sj26GST-SjFABP significantly increased levels of antibody, splenocyte proliferative response and production of IFN-gamma, compared with rPRV/Sj26GST and rPRV/SjFABP. These recombinant viruses have been shown to be safe for sheep. Challenge experiments showed worms and egg burdens were significantly reduced in animals immunized with recombinant PRVs. Most importantly, rPRV/Sj26GST-SjFABP dramatically enhanced protection with worm reduction and hepatic reduction of 39.3% and 45.5% respectively in mice, and 48.5% and 51.2% in sheep, while rPRV/Sj26GST and rPRV/SjFABP provided corresponding protection of only up to 23.7% and 27.2% in mice, and 29.0% and 35.5% in sheep. These results indicate that the multivalent vaccine for S. japonicum can produce significant specific immunity and protection, and that PRV Bartha-K61 is an effective live vector for an animal schistosomiasis japonica vaccine.
Collapse
Affiliation(s)
- Feng Wei
- Institute of Military Veterinary, AMMS, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun 130062, Jilin Province, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Wu WC, Jin DY, Lou WH, Wang DS, Qin XY. Induction of antigen-specific CTL and antibody responses in mice by a novel recombinant tandem repeat DNA vaccine targeting at mucin 1 of pancreatic cancer. J Cancer Res Clin Oncol 2010; 136:1861-8. [PMID: 20229033 DOI: 10.1007/s00432-010-0845-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2010] [Accepted: 02/15/2010] [Indexed: 01/22/2023]
Abstract
PURPOSE Tandem repeat (TR) is the key epitope of mucin 1 (MUC1) for inducing cytotoxic T lymphocytes (CTL) to kill the tumor cells specifically. This study aimed to construct a new recombinant DNA vaccine based on single TR and to investigate the induced immune responses in mice. MATERIALS AND METHODS After the synthesis of a recombinant human TR(rhTR)and the construction of the recombinant plasmid pcDNA3.1-TR/Myc-his (+) A (pTR plasmid), C57BL/6 (H-2(b)) mice were immunized with it (TR group, n = 15). Mice inoculated with the empty vector (EV group, n = 15) and normal saline (NS group, n = 15) were used as vector and blank control, respectively. Cytotoxic assay was carried out to measure the CTL activity. And indirect enzyme-linked immunosorbent assay (ELISA) was used to detect anti-TR-specific antibodies. RESULTS TR group resulted in more efficient induction of CTL-specific cytolysis against TR polypeptide than both EV and NS groups (both P < 0.01). Vaccine-immunized mice had a higher equivalent concentration of anti-TR-specific antibodies (2,324 ± 238 μg/ml) than either of EV group (1,896 ± 533 μg/ml, P < 0.01) or NS group (1,736 ± 142 μg/ml, P < 0.01). CONCLUSION The novel recombinant TR DNA vaccine targeting at MUC1 of pancreatic cancer was constructed successfully, effectively expressing TR polypeptide in the transfected mammalian cells and inducing TR-specific CTL and antibody response.
Collapse
Affiliation(s)
- Wen-Chuan Wu
- Department of General Surgery, Zhongshan Hospital, Fudan University, 180 Feng Lin Road, Shanghai 200032, People's Republic of China
| | | | | | | | | |
Collapse
|
21
|
Immune responses to polyethylenimine-mannose-delivered plasmid DNA encoding a Fasciola gigantica fatty acid binding protein in mice. J Helminthol 2009; 84:149-55. [PMID: 19723356 DOI: 10.1017/s0022149x0999037x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Fasciola gigantica fatty acid binding protein (FABP) was evaluated for evoking an immune response in mice, by delivering the gene coding for this protein with mannosylated-polyethylenimine (PEI) to peritoneal cells. Mice were immunized with 50 microg recombinant plasmid DNA (Group I) or DNA-PEI-mannose (a 22 kDa linear cationic polymer with mannose ligand) (Group II) via the intraperitoneal route. Antibody studies showed no significant humoral immune response evoked to this DNA immunization with either PEI-mannose-delivered or naked DNA. However, on protein boosting of these DNA-primed mice there was a significant enhancement of antibody titre. Flow cytometric bead array was used to measure quantities of interleukin (IL)-2, IL-4, IL-5, interferon-gamma (IFN-gamma) and tumour necrosis factor (TNF) cytokines. Overexpression of T-helper 1 (Th1) cytokines such as IFN-gamma and TNF, with a lower but significant expression of the T-helper 2 (Th2) cytokine IL-5 was detected. Gene delivery using polyethylenimine-mannose ligand showed significant expression of IFN-gamma and TNF (P < 0.05), but no significant difference in IL-2, IL-4 and IL-5 (P>0.05) cytokine expression was observed between naked-DNA- and mannosylated PEI-DNA-delivered mice. Naked- or PEI-delivered-DNA immunization produced insignificant levels of IL-2 and IL-4 (P>0.05) cytokines in both groups of mice.
Collapse
|
22
|
Wei F, Liu Q, Zhai Y, Fu Z, Liu W, Shang L, Men J, Gao S, Lian H, Jin H, Chen C, Lin J, Shi Y, Xia Z, Zhu XQ. IL-18 enhances protective effect in mice immunized with a Schistosoma japonicum FABP DNA vaccine. Acta Trop 2009; 111:284-8. [PMID: 19467215 DOI: 10.1016/j.actatropica.2009.03.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2008] [Revised: 03/13/2009] [Accepted: 03/24/2009] [Indexed: 12/16/2022]
Abstract
Two recombinant plasmids, pVAX/SjFABP and pVAX/mIL-18 containing Schistosoma japonicum 14 kDa fatty acid binding protein (SjFABP) and murine IL-18, were constructed and evaluated for their ability to induce immune responses and to protect against S. japonicum challenge in mice. Mice were intramuscularly immunized twice at three-weekly intervals, and challenged with S. japonicum cercariae at 4 weeks after the last vaccination. All animals vaccinated with pVAX/SjFABP alone or plus pVAX/mIL-18 developed specific anti-SWAP ELISA antibody and T lymphocyte proliferation. Co-injection of pVAX/mIL-18 significantly increased the production of IFN-gamma and IL-2 compared with pVAX/SjFABP alone, indicating that IL-18 enhances the Th1-dominant immune response. The challenge experiment showed that co-injection of plasmid encoding IL-18 significantly enhances protective effect against S. japonicum infection, as demonstrated by worm reduction rates and the hepatic egg reduction rates 45 days post-challenge. These results indicated that IL-18 may become a novel vaccine adjuvant for development of vaccines against schistosomiasis.
Collapse
|
23
|
Adult Brugia malayi approximately 34 kDa (BMT-5) antigen offers Th1 mediated significant protection against infective larval challenge in Mastomys coucha. Parasitol Int 2009; 58:346-53. [PMID: 19638321 DOI: 10.1016/j.parint.2009.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2009] [Revised: 07/08/2009] [Accepted: 07/10/2009] [Indexed: 10/20/2022]
Abstract
We earlier reported a sizeable protection conferred by 'mitochondria rich' (MT) fraction of adult B. malayi and the present study was planned to locate the candidate protective molecule/s in the active fraction. The MT fraction was subjected to sodium dodecyl sulphate polyacrylamide gel electrophoresis (SDS-PAGE) and the antigen bands showing strong immune-reactivity with the resistant mastomys sera were assayed for their lymphoproliferative response using splenocytes of protected animals. Of the eight such protein bands, one sub fraction with a molecular weight of approximately 34 kDa (BMT-5) produced utmost cellular proliferation and was therefore exploited for vaccination study. BMT-5 emulsified in Freund's adjuvant produced discernible protection causing 69 and 67% reductions in microfilaraemia and adult worm burden respectively along with sterilization of 68% of the recovered female parasites. Significant levels of filaria-specific and non-specific lymphoproliferation along with enhanced release of Th1 cytokines (TNF-alpha, IFN-gamma and IL-2) by splenocytes were observed in the vaccinated mastomys in addition to elevated levels of antigen-specific IgG, IgG2a, IgG2b and IgA. The peritoneal macrophages of immunized animals also revealed enhanced nitric oxide production in the presence of BMT-5. The findings suggest that approximately 34 kDa (BMT-5) molecules present in the MT fraction of adult B. malayi provided sizeable protection against infective larval challenge by generating a Th1 biased milieu in the host.
Collapse
|
24
|
Th1-type epitopes-based cocktail PDDV attenuates hepatic fibrosis in C57BL/6 mice with chronic Schistosoma japonicum infection. Vaccine 2009; 27:4110-7. [PMID: 19410625 DOI: 10.1016/j.vaccine.2009.04.073] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Revised: 04/20/2009] [Accepted: 04/25/2009] [Indexed: 01/15/2023]
Abstract
Schistosomiasis is one of the world's major public health problems in terms of morbidity and mortality, which is characterized by a marked egg-induced CD4(+) T-cell programmed granulomatous inflammation and cumulative fibrosis. Here PDDV (peptide-DNA dual vaccine), a widely used non-viral gene delivery system, was applied. The cocktail PDDV, based on four Th1-type epitope peptides identified from Schistosoma japonicum vaccine candidates and CpG ODN1826, could induce dominant Th1-type response in C57BL/6J mice (P<0.05). The histopathological staging and collagen assessment for fibrosis showed that the cocktail PDDV presented an obvious down-regulation effect on hepatic fibrosis caused by chronic S. japonicum infection (P<0.05), and IFN-gamma, IL-4 and IL-13 mRNAs in liver detected by RT-PCR also showed that the cocktail PDDV represented the ability to up-regulate Th1-type responses, which paralleled with a decrease expression of alpha-SMA (P<0.05) and the up-regulated MMP9/TIMP1 balance (P<0.05) when compared to the control groups. Therefore, it is indicated that the cocktail PDDV can significantly attenuate hepatic fibrosis, in parallel with the decreased HSCs activation and the up-regulated MMP9/TIMP1 balance in favor of matrix degradation, which may be partially dependent on the increased Th1 response to restore the Th1/Th2 balance.
Collapse
|
25
|
Anand SB, Murugan V, Prabhu PR, Anandharaman V, Reddy MVR, Kaliraj P. Comparison of immunogenicity, protective efficacy of single and cocktail DNA vaccine of Brugia malayi abundant larval transcript (ALT-2) and thioredoxin peroxidase (TPX) in mice. Acta Trop 2008; 107:106-12. [PMID: 18547532 DOI: 10.1016/j.actatropica.2008.04.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2007] [Revised: 04/18/2008] [Accepted: 04/22/2008] [Indexed: 11/30/2022]
Abstract
Although DNA vaccines have several advantages over conventional vaccines, antibody production and protection are often not adequate, particularly in single plasmid vaccine formulation. In the present study we evaluated the efficacy of a cocktail vaccine based on plasmids encoding larval (L3) stage-specific Brugia malayi abundant larval transcript (BmALT-2) and antioxidant detoxification enzyme B. malayi thioredoxin peroxidase (BmTPX) to induce antibodies, protective efficacy and cell-mediated immune response in mice. Mice immunized with cocktail DNA vaccines containing the pVAX ALT-2+TPX developed higher titers of anti-BmALT-2+TPX (1:5000) antibodies, compared to the mice immunized with single DNA vaccine of pVAX ALT-2 or pVAX TPX (1:2000). Correlating with this, the mice administered with cocktail vaccine induced up to 78% of cytotoxicity against B. malayi mf. This cytotoxicity was high compared to 34% induced by the pVAX-ALT2 or 37% by pVAX-TPX. Moreover, cocktail vaccination of mice resulted in significantly higher level of cellular proliferative response associated with raised levels of IFN-gamma that skewed towards Th1 type of response compared to vaccination using either of the components. Taken together, these data suggest that the combination of two or more antigens maybe an effective vaccine development strategy to improve protection and immunogenicity against human lymphatic filariasis.
Collapse
|
26
|
Wei F, Liu Q, Gao S, Shang L, Zhai Y, Men J, Jiang L, Zhu XQ, Fu Z, Shi Y, Xia Z, Lin J. Enhancement by IL-18 of the protective effect of a Schistosoma japonicum 26kDa GST plasmid DNA vaccine in mice. Vaccine 2008; 26:4145-9. [PMID: 18562051 DOI: 10.1016/j.vaccine.2008.05.034] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2008] [Revised: 05/15/2008] [Accepted: 05/15/2008] [Indexed: 01/10/2023]
Abstract
Two recombinant plasmids pVAX/Sj26GST and pVAX/mIL-18 containing Schistosoma japonicum 26kDa GST and murine IL-18 were evaluated for their ability to protect mice against S. japonicum challenge. Mice were given 2 intramuscular immunizations 3 weeks apart, and challenged with S. japonicum cercariae 4 weeks later. Adult worm and egg burdens were determined 48 days post-challenge. All animals vaccinated with pVAX/Sj26GST alone or with pVAX/mIL-18 developed specific anti-SWAP (soluble worm antigen preparation) ELISA antibody and splenocyte proliferation response. Co-injection of pVAX/mIL-18 significantly increased the production of IFN-gamma and IL-12, indicating that IL-18 enhances the Th1-dominant immune response. Challenge experiments showed that worms were reduced in the pVAX/Sj26GST group by 30.1% and by 49.4% in animals given pVAX/mIL-18 additionally. Corresponding hepatic and fecal egg reductions were 44.8% and 53.0%, and 50.6% and 56.6%, respectively. These results indicate that IL-18 may be an effective adjuvant for a schistosomiasis vaccine.
Collapse
Affiliation(s)
- Feng Wei
- Laboratory of Parasitology, Veterinary Institute, Academy of Military Medical Sciences, 1068 Qinglong Road, Changchun 130062, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Immunization of mice with cells from juvenile worms of Schistosoma japonicum provides immunoprotection against schistosomiasis. ACTA ACUST UNITED AC 2008; 50:822-30. [PMID: 17973094 DOI: 10.1007/s11427-007-0100-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2007] [Accepted: 08/26/2007] [Indexed: 10/22/2022]
Abstract
To validate the protective efficacy against schistosomiasis by immunization with cells from juvenile Schistosoma japonicum in a murine model and to analyze possible factors related to protection, in this study, two independent repeated vaccination trials were performed. After three subcutaneous vaccinations, in trial one, in the absence of adjuvant, primary juvenile worm cells (pJCs) from S. japonicum induced remarkable average reductions in worm burden (54.3%), liver eggs per gram (LEPG) load (59.8%) as well as egg granulomas size (66.5%) compared to PBS control group (P<0.01), which were significantly higher than those elicited by fractions of juvenile worm cells (JCFs) or fractions of juvenile worms (JWFs) (P<0.05). Non-cell components of worms (WNCs) showed no significant protection. In trial two, compared to PBS control group, significant protective effect was also observed for cultured juvenile worm cells (cJCs) from S. japonicum with 58.4% worm reduction and 68.1% LEPG reduction (P<0.01). However, cultured adult worms cells (cACs) showed significantly higher worm burden (P<0.05) and egg burden (P<0.01) when compared to cJCs. Immunological analysis of trial two revealed that cJCs engendered a Th1-biased mixed Th1/Th2 type of immune response while cACs elicited a Th2-type response. Our data indicated that immunization with both primary and cultured cells from S. japonicum juvenile worms provided high immunoprotection, for which the physical character of immunogens, stage-specific parasite and the type of immune response induced might be responsible, suggesting that vaccination with whole cells from S. japonicum larvae is a promising approach to produce protective immunity against schistosomiasis.
Collapse
|
28
|
Gottstein B, Hemphill A. Echinococcus multilocularis: the parasite-host interplay. Exp Parasitol 2008; 119:447-452. [PMID: 18410929 DOI: 10.1016/j.exppara.2008.03.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2007] [Revised: 02/27/2008] [Accepted: 03/06/2008] [Indexed: 11/16/2022]
Abstract
Alveolar echinococcosis (AE) is a severe chronic helminthic disease caused by the intrahepatic tumor-like growth of the metacestode of Echinococcus multilocularis. Metacestodes are fluid-filled, asexually proliferating vesicles, which are entirely covered by the laminated layer, an acellular carbohydrate-rich surface structure that protects the parasite from immunological and physiological reactions on part of the host. The E. multilocularis metacestode has acquired specific means of manipulating and using the immunological host response to its own advantage. These include the expression of distinct immunoregulatory parasite molecules that manipulate and interfere in the functional activity of macrophages and T cells. Recent research findings have led to a better understanding of the protein- and glycoprotein composition of the laminated layer and the E/S fraction of the metacestode, including Em2- and Em492-antigens, two metacestode antigen fractions that exhibit immunosuppressive or -modulatory properties. Understanding of the events taking place at the host-parasite interface is the key for development of novel immuno-therapeutical and/or chemotherapeutical tools.
Collapse
Affiliation(s)
- Bruno Gottstein
- Institute of Parasitology, Vetsuisse Faculty and Faculty of Medicine, University of Bern, Laenggassstrasse 122, CH-3001 Bern, Switzerland.
| | | |
Collapse
|
29
|
Siles-Lucas M, Uribe N, López-Abán J, Vicente B, Orfao A, Nogal-Ruiz JJ, Feliciano AS, Muro A. The Schistosoma bovis Sb14-3-3ζ recombinant protein cross-protects against Schistosoma mansoni in BALB/c mice. Vaccine 2007; 25:7217-23. [PMID: 17707955 DOI: 10.1016/j.vaccine.2007.07.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2006] [Revised: 05/14/2007] [Accepted: 07/15/2007] [Indexed: 11/19/2022]
Abstract
Current control programs against schistosomiasis could be reinforced through the use of an effective vaccine. Schistosome 14-3-3 proteins have been proposed as candidates for vaccine against the respective infections, and were seen to elicit high protection levels against Schistosoma bovis in a previous work done by our group. We have therefore investigated the protective capacity of the 14-3-3 protein from S. bovis - Sb14zeta - against Schistosoma mansoni in mice. In addition, we have addressed the influence of the co-administration of three different immunomodulators with the 14-3-3 polypeptide. Protection was high when the Sb14zeta protein was combined in two independent experiments with the AA2829 and PAL immunomodulatory molecules as regards both the reduction of worm numbers (mean: 64.8%) and egg loads in liver (mean: 73.9%) or intestine (mean: 71.5%). In contrast, the degree of protection achieved with the Sb14zeta-CpG vaccine was very low (14.9% reduction in worm numbers, and 46.6% and 32% reduction in liver and intestinal egg loads). The immune responses observed in the vaccinated animals showed that the production of IFNgamma and the absence of IL-4, accompanied by a strong humoral response, are insufficient to elicit protection against S. mansoni.
Collapse
Affiliation(s)
- M Siles-Lucas
- Laboratorio de Parasitología, Facultad de Farmacia, CIETUS, Universidad de Salamanca, Avda. Campo Charro, s/n. 37007 Salamanca, Spain
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Monroy FP. Toxoplasma gondii: effect of infection on expression of 14-3-3 proteins in human epithelial cells. Exp Parasitol 2007; 118:134-8. [PMID: 17825295 PMCID: PMC2244797 DOI: 10.1016/j.exppara.2007.07.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Revised: 06/26/2007] [Accepted: 07/16/2007] [Indexed: 11/21/2022]
Abstract
14-3-3 Proteins are expressed in most eukaryotes organisms and play varied and crucial roles in a wide range of regulatory processes. In mammalian cells, seven 14-3-3 isoforms have been identified. However, it is not known what effect infection has on 14-3-3 isoform expression. In this study human colonic carcinoma cell lines were infected with Toxoplasma gondii for 24h and expression of 14-3-3 proteins was determined by RT-PCR. HT-29 cells only expressed 3 out of the 7 isoforms while 5 and all 7 isoforms were found in HCT-116 and Caco-2 cells, respectively. Infection had little or no effect in the expression of 14-3-3gamma, epsilon, sigma, and xi; but in HCT-116 cells induced expression of 14-3-3eta and sigma, while 14-3-3beta, eta, and xi were induced in HT-29 cells. If 14-3-3 proteins are involved in cell survival and/or prevention of parasite replication, longer incubation times may be required as no differences in percentage of infection were found among the cell lines at 24h post-infection.
Collapse
Affiliation(s)
- Fernando P Monroy
- Department of Biological Sciences, Northern Arizona University, PO Box 5640, Flagstaff, AZ 86011, USA.
| |
Collapse
|
31
|
Uribe N, Muro A, Vieira C, Lopez-Aban J, del Olmo E, Suárez L, Martínez-Fernández AR, Siles-Lucas M. Genetic and Immunological Characterization of the 14-3-3ζ Molecule From Schistosoma bovis. J Parasitol 2007; 93:964-9. [DOI: 10.1645/ge-999r2.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
32
|
Chen AY, Fry SR, Forbes-Faulkner J, Daggard G, Mukkur TKS. Evaluation of the immunogenicity of the P97R1 adhesin of Mycoplasma hyopneumoniae as a mucosal vaccine in mice. J Med Microbiol 2006; 55:923-929. [PMID: 16772421 DOI: 10.1099/jmm.0.46088-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The immunogenicity of P97 adhesin repeat region R1 (P97R1) of Mycoplasma hyopneumoniae, an important pathogenesis-associated region of P97, was evaluated in mice as a mucosal vaccine. Mice were immunized orally with attenuated Salmonella typhimurium aroA strain CS332 harbouring a eukaryotic or prokaryotic expression vector encoding P97R1. Local and systemic immune responses were analysed by ELISA on mouse sera, lung washes and splenocyte supernatants following splenocyte stimulation with specific antigens in vitro. Although no P97R1-specific antibody responses were detected in serum and lung washes, significant gamma interferon was produced by P97R1-stimulated splenocytes from mice immunized orally with S. typhimurium aroA harbouring either expression system, indicating induction of a cell-mediated immune response. These results suggested that live bacterial vectors carrying DNA vaccines or expressing heterologous antigens preferentially induce a Th1 response. Surprisingly, however, mice immunized with the vaccine carrier S. typhimurium aroA CS332 induced serum IgG, but not mucosal IgA, against P97R1 or S. typhimurium aroA CS332 whole-cell lysate, emphasizing the importance of assessing the suitability of attenuated S. typhimurium antigen-carrier delivery vectors in the mouse model prior to their evaluation as potential vaccines in the target species, which in this instance was pigs.
Collapse
MESH Headings
- Adhesins, Bacterial/genetics
- Adhesins, Bacterial/immunology
- Amino Acid Sequence
- Animals
- Antibodies, Bacterial/blood
- Bacterial Vaccines/genetics
- Bacterial Vaccines/immunology
- Base Sequence
- DNA, Bacterial/chemistry
- DNA, Bacterial/genetics
- Female
- Interferon-gamma/immunology
- Mice
- Mice, Inbred BALB C
- Molecular Sequence Data
- Mycoplasma hyopneumoniae/genetics
- Mycoplasma hyopneumoniae/immunology
- Pneumonia of Swine, Mycoplasmal/immunology
- Pneumonia of Swine, Mycoplasmal/microbiology
- Pneumonia of Swine, Mycoplasmal/prevention & control
- Polymerase Chain Reaction
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Salmonella typhimurium/genetics
- Spleen/immunology
- Spleen/virology
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Austen Y Chen
- Department of Biological and Physical Sciences, University of Southern Queensland, Toowoomba, Queensland, Australia
| | - Scott R Fry
- Department of Biological and Physical Sciences, University of Southern Queensland, Toowoomba, Queensland, Australia
| | | | - Grant Daggard
- Department of Biological and Physical Sciences, University of Southern Queensland, Toowoomba, Queensland, Australia
| | - T K S Mukkur
- Department of Biological and Physical Sciences, University of Southern Queensland, Toowoomba, Queensland, Australia
| |
Collapse
|
33
|
Ganley-Leal LM, Guarner J, Todd CW, Da'Dara AA, Freeman GL, Boyer AE, Harn DA, Secor WE. Comparison of Schistosoma mansoni irradiated cercariae and Sm23 DNA vaccines. Parasite Immunol 2005; 27:341-9. [PMID: 16149992 DOI: 10.1111/j.1365-3024.2005.00785.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Immunization with defined antigens is generally less effective at inducing host protection against experimental infection with Schistosoma mansoni than vaccination with attenuated infective cercariae. We predicted that quantitative and/or qualitative differences existed between the immune responses generated to attenuated cercariae and those induced by defined antigens. Thus, we compared immune responses typically associated with protection in the murine model between animals vaccinated with attenuated cercariae and mice immunized with DNA encoding Sm23, a schistosome integral membrane protein that has previously been shown to confer protection. Mice vaccinated three times with attenuated cercariae demonstrated higher levels of protection than Sm23-vaccinated animals but spleen cells from Sm23 DNA vaccinated mice produced significantly higher levels of schistosome antigen-specific IFN-gamma. Both vaccines induced similar levels of Sm23-specific antibody and post-challenge dermal inflammation. However, the pulmonary inflammatory responses following challenge were much less pronounced in DNA immunized animals compared to those receiving irradiated cercariae. Thus, although Sm23 DNA vaccination effectively induced parasite-specific IFN-gamma and antibody responses, it failed to evoke other critical responses needed for optimal vaccine efficacy.
Collapse
Affiliation(s)
- L M Ganley-Leal
- Division of Parasitic Diseases, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia 30341, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
McManus DP. Prospects for development of a transmission blocking vaccine against Schistosoma japonicum. Parasite Immunol 2005; 27:297-308. [PMID: 16138851 DOI: 10.1111/j.1365-3024.2005.00784.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Despite intensive long-term control programmes, schistosomiasis japonica remains a serious public health problem in China and the Philippines. The termination of mass praziquantel-treatment has seen a dramatic recent rebound in both its prevalence and associated morbidity. Schistosomiasis japonica is a zoonosis but, despite complicating control efforts, this feature provides a practical method for attacking Schistosoma japonicum through development and deployment of a transmission blocking veterinary vaccine. A recently completed bovine drug intervention trial and mathematical modelling of the transmission of S. japonicum underpin the concept that such a vaccine, targeting water buffalo, would have major implications for future integrated schistosomiasis control in China. A major block to success is the low ceiling efficacy achieved with current vaccine molecules. To solve this challenge, an antigen discovery pipeline needs to be established for identification of new vaccine targets that induce greater potency than the current anti-S. japonicum candidate vaccines. Excretory-secretory products and molecules exposed on epithelial surfaces (including receptors) which interact directly with the host immune system warrant especial attention. Extensive schistosome genomics programmes currently underway coupled with new advances in proteomics and microarray technology provide an unparalleled opportunity to identify new molecules exploitable as vaccine targets. These will then need to be produced in quantity and rigorously tested first in the laboratory and then the field. If a transmission blocking veterinary vaccine developed for bovines can be put into practice in combination with other control strategies such as human chemotherapy, elimination of S. japonicum from China may be achievable.
Collapse
Affiliation(s)
- D P McManus
- Molecular Parasitology Laboratory, Queensland Institute of Medical Research, Brisbane, Australia.
| |
Collapse
|
35
|
Abstract
This is the first of a projected series of canonic reviews covering all invertebrate muscle literature prior to 2005 and covers muscle genes and proteins except those involved in excitation-contraction coupling (e.g., the ryanodine receptor) and those forming ligand- and voltage-dependent channels. Two themes are of primary importance. The first is the evolutionary antiquity of muscle proteins. Actin, myosin, and tropomyosin (at least, the presence of other muscle proteins in these organisms has not been examined) exist in muscle-like cells in Radiata, and almost all muscle proteins are present across Bilateria, implying that the first Bilaterian had a complete, or near-complete, complement of present-day muscle proteins. The second is the extraordinary diversity of protein isoforms and genetic mechanisms for producing them. This rich diversity suggests that studying invertebrate muscle proteins and genes can be usefully applied to resolve phylogenetic relationships and to understand protein assembly coevolution. Fully achieving these goals, however, will require examination of a much broader range of species than has been heretofore performed.
Collapse
Affiliation(s)
- Scott L Hooper
- Neuroscience Program, Department of Biological Sciences, Irvine Hall, Ohio University, Athens, Ohio 45701, USA.
| | | |
Collapse
|
36
|
Kerepesi LA, Keiser PB, Nolan TJ, Schad GA, Abraham D, Nutman TB. DNA immunization with Na+-K+ ATPase (Sseat-6) induces protective immunity to larval Strongyloides stercoralis in mice. Infect Immun 2005; 73:2298-305. [PMID: 15784574 PMCID: PMC1087441 DOI: 10.1128/iai.73.4.2298-2305.2005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2004] [Revised: 10/25/2004] [Accepted: 11/26/2004] [Indexed: 11/20/2022] Open
Abstract
Strongyloides stercoralis causes chronic asymptomatic infections which can be maintained in the human host for many decades. Identification and treatment of S. stercoralis-infected individuals is required because immunosuppression can lead to fatal hyperinfection. In this study, human immunoglobulin G (IgG) that had previously been shown to transfer protective immunity to mice was used to identify potential protective antigens. Three antigens or genes from S. stercoralis larvae were identified as tropomyosin (Sstmy-1), Na+-K+ ATPase (Sseat-6), and LEC-5 (Sslec-5). The genes were cloned into plasmids for DNA immunization, and mice were immunized intradermally with the three plasmids individually in combination with a plasmid containing murine granulocyte-macrophage colony-stimulating factor. Only Na+-K+ ATPase induced a significant reduction in larval survival after DNA immunization. Immunization with a combination of all three plasmids, including Na+-K+ ATPase, did not induce protective immunity. Serum from mice immunized with DNA encoding Na+-K+ ATPase was transferred to naive mice and resulted in partial protective immunity. Therefore, DNA immunization with Na+-K+ ATPase induces protective immunity in mice, and it is the first identified vaccine candidate against infection with larval S. stercoralis.
Collapse
Affiliation(s)
- Laura A Kerepesi
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | |
Collapse
|
37
|
Siddiqui AA, Pinkston JR, Quinlin ML, Saeed Q, White GL, Shearer MH, Kennedy RC. Characterization of the immune response to DNA vaccination strategies for schistosomiasis candidate antigen, Sm-p80 in the baboon. Vaccine 2005; 23:1451-6. [PMID: 15670880 DOI: 10.1016/j.vaccine.2004.09.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2004] [Revised: 08/27/2004] [Accepted: 09/28/2004] [Indexed: 10/26/2022]
Abstract
Even though schistosomicidal agents and other control measures, including public hygiene and snail control exist, development of an efficacious vaccine still remains the most potentially powerful method for control of schistosomiasis. In our continuing efforts to develop a vaccine against schistosomiasis, we have selected a vaccine candidate (Sm-p80), which plays an important role in the immune evasion process of the parasite. Sm-p80 has been shown to confer up to 60% protection in mice following experimental infection. In this initial study, we have used Sm-p80 plus the Th1 response promoting cytokine, interleukin-2 (IL-2), in a DNA immunogen formulation. The vaccine was tested for its safety and immunogenicity in a baboon model of schistosomiasis. The vaccine generated a Th1 type Sm-p80-specific response in baboons with IgG(1)/IgG(2) ratios of less than 1.0. No detectable IgG(3) or IgG(4) anti-Sm-p80 responses were present in the immunized baboons. The antibodies to Sm-p80 were able to kill up to 35% schistosomula in vitro in the presence of complement. These results although preliminary suggest the potential of Sm-p80 as a viable vaccine candidate for schistosomiasis.
Collapse
Affiliation(s)
- Afzal A Siddiqui
- Department of Internal Medicine, Texas Tech Women's Health and Research Institute, 1400 Wallace Blvd., Amarillo, TX 79106-1791, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Erttmann KD, Kleensang A, Schneider E, Hammerschmidt S, Büttner DW, Gallin M. Cloning, characterization and DNA immunization of an Onchocerca volvulus glyceraldehyde-3-phosphate dehydrogenase (Ov-GAPDH). Biochim Biophys Acta Mol Basis Dis 2005; 1741:85-94. [PMID: 15955451 DOI: 10.1016/j.bbadis.2004.12.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2004] [Revised: 12/05/2004] [Accepted: 12/14/2004] [Indexed: 11/18/2022]
Abstract
In the search for Onchocerca volvulus antigens possibly involved in protection against human onchocerciasis, partial amino acid sequence analysis of one of the O. volvulus antigens of the serologically identified proteins showed a close relationship to the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) protein family. Subsequent adult worm cDNA library screening and cloning produced a clone of 1650 bp. An open reading frame spans over 1020 bp encoding for a protein of 340 amino acids with an apparent molecular weight of 38000. Comparison of the complete amino acid sequence identified this protein as a member of the GAPDH protein family. The recombinantly expressed protein shows GAPDH enzymatic activity as well as plasminogen-binding capacity. DNA sequence analysis of the corresponding gene revealed the presence of two introns. Using immunohistology Ov-GAPDH was observed in microfilariae, infective larvae, and adult male and female worms. Most striking was the labelling of the musculature of the body wall. Labelling was also observed in the pseudocoeloma cavity and in a subset of cell nuclei, suggesting additional, non-glycolytic functions of the Ov-GAPDH. Gene gun immunization with the DNA-construct in cattle led to specific humoral immune responses. Thus, the protective potential of the DNA-construct of Ov-GAPDH can be evaluated in vaccination trials using animal models such as the cattle/Onchocerca ochengi model.
Collapse
Affiliation(s)
- Klaus D Erttmann
- Bernhard Nocht Institute for Tropical Medecine, Bernhard-Nocht-Str.74, D-20359 Hamburg, Germany.
| | | | | | | | | | | |
Collapse
|
39
|
Ivory C, Chadee K. DNA vaccines: designing strategies against parasitic infections. GENETIC VACCINES AND THERAPY 2004; 2:17. [PMID: 15579202 PMCID: PMC544584 DOI: 10.1186/1479-0556-2-17] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/05/2004] [Accepted: 12/03/2004] [Indexed: 11/28/2022]
Abstract
The complexity of parasitic infections requires novel approaches to vaccine design. The versatility of DNA vaccination provides new perspectives. This review discusses the use of prime-boost immunizations, genetic adjuvants, multivalent vaccines and codon optimization for optimal DNA vaccine design against parasites.
Collapse
Affiliation(s)
- Catherine Ivory
- Institute of Parasitology of McGill University, Macdonald Campus, 21,111 Lakeshore Road, Ste. Anne de Bellevue, Quebec, Canada, H9X 3V9
| | - Kris Chadee
- Institute of Parasitology of McGill University, Macdonald Campus, 21,111 Lakeshore Road, Ste. Anne de Bellevue, Quebec, Canada, H9X 3V9
| |
Collapse
|
40
|
Jones MK, Gobert GN, Zhang L, Sunderland P, McManus DP. The cytoskeleton and motor proteins of human schistosomes and their roles in surface maintenance and host-parasite interactions. Bioessays 2004; 26:752-65. [PMID: 15221857 DOI: 10.1002/bies.20058] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Schistosomes are parasitic blood flukes, responsible for significant human disease in tropical and developing nations. Here we review information on the organization of the cytoskeleton and associated motor proteins of schistosomes, with particular reference to the organization of the syncytial tegument, a unique cellular adaptation of these and other neodermatan flatworms. Extensive EST databases show that the molecular constituents of the cytoskeleton and associated molecular systems are likely to be similar to those of other eukaryotes, although there are potentially some molecules unique to schistosomes and platyhelminths. The biology of some components, particular those contributing to host-parasite interactions as well as chemotherapy and immunotherapy are discussed. Unresolved questions in relation to the structure and function of the tegument relate to dynamic organization of the syncytial layer.
Collapse
Affiliation(s)
- Malcolm K Jones
- Queensland Institute of Medical Research, Herston, Queensland, Australia.
| | | | | | | | | |
Collapse
|
41
|
Abstract
There is continued transmission of schistosomiasis japonica in China and Philippines despite highly effective control programs that focus on the application of the highly effective drug praziquantel (PZQ). The massive Three Gorges Dam across the Yangtze River in Southern China, soon to be completed, is expected to significantly increase schistosomiasis transmission and introduce the disease into areas currently unaffected. After long-term experience it is generally accepted that PZQ chemotherapy, although the cornerstone of current control programs, does have significant limitations. Furthermore, efficient drug delivery requires a substantial infrastructure to regularly cover all parts of an endemic area. Although there is not yet clear-cut evidence for the existence of PZQ-resistant schistosome strains, decreased susceptibility to the drug has been observed in several countries. As a result, a protective vaccine represents an essential component for the long-term control of schistosomiasis. This article briefly reviews aspects of anti-schistosome protective immunity that are important in the context of vaccine development. The current status in the development of vaccines against Schistosoma japonicum will then be discussed as will new approaches that may improve on the efficacy of available vaccines, and aid in the identification of new targets for immune attack. With new and extensive data becoming available from the S. japonicum genome project, the prospects for developing an effective vaccine are encouraging. The challenges that remain are many but it is crucial that the momentum towards developing effective anti-schistosome vaccines is maintained.
Collapse
Affiliation(s)
- Donald P McManus
- Molecular Parasitology Laboratory, Australian Center for International and Tropical Health and Nutrition, Queensland Institute of Medical Research, 300 Herston Road, Brisbane, QLD 4029, Australia.
| | | |
Collapse
|
42
|
Singh A, Singh A, Chaudhri SS. Visceral schistosomiasis of domestic animals in India: humoral immune status of infected cattle, sheep and goats against major polypeptide antigens of Schistosoma indicum and S. spindale. Parasite Immunol 2004; 26:167-75. [PMID: 15367294 DOI: 10.1111/j.0141-9838.2004.00697.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Polypeptide profiles of Schistosoma indicum and S. spindale adult worm homogenates were obtained by sodium dodecyl sulphate-polyacrylamide gel electrophoresis. Humoral immune status of infected cattle, sheep and goats against Schistosoma indicum and S. spindale Ags was determined by immunoblot analysis and by indirect ELISA using four major polypeptides of approximate molecular masses 45 kDa, 40 kDa, 28 kDa and 15 kDa electro-eluted from the gel slices. Cattle sera samples had higher levels of antibodies against Si/s40 and Si/s28 than against Si/s45 antigen. Reasons have been discussed for the absence of detectable levels of anti-Si/s28, -Si/s45 and -Si/s40 antibodies in a significant number of sera samples from S. indicum egg-positive sheep.
Collapse
Affiliation(s)
- A Singh
- Immunology Section, Department of Veterinary Microbiology, Ch. Charan Singh Haryana Agricultural University, Hisar (Haryana), India
| | | | | |
Collapse
|
43
|
Siddiqui AA, Phillips T, Charest H, Podesta RB, Quinlin ML, Pinkston JR, Lloyd JD, Paz M, Villalovos RM, Pompa J. Induction of protective immunity against Schistosoma mansoni via DNA priming and boosting with the large subunit of calpain (Sm-p80): adjuvant effects of granulocyte-macrophage colony-stimulating factor and interleukin-4. Infect Immun 2003; 71:3844-51. [PMID: 12819068 PMCID: PMC161986 DOI: 10.1128/iai.71.7.3844-3851.2003] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Considerable morbidity and mortality result from schistosomiasis, an affliction affecting an estimated 200 million people. Although schistosomicidal drugs and other control measures (including public hygiene and snail control) exist, the advent of an efficacious vaccine remains the most potentially powerful means for controlling this disease. We have targeted a vaccine candidate (large subunit of calpain, Sm-p80) because of its consistent immunogenicity, protective potential, and integral role in surface membrane biogenesis of schistosomes. Since surface membrane renewal appears to be one of the major phenomena employed by schistosomes to evade the host's immune system; an immune response directed against Sm-p80 should render the parasite susceptible to immune clearance from the host by both providing a focus of attack and by potentially impairing the membrane repair process. In the present study, we have employed DNA immunization protocols using Sm-p80 with plasmids encoding granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL-4). Sm-p80 by itself provided 39% protection (P = < or =0.0001) against challenge infection in C57BL/6 mice. This protection was increased to 44% (P = < or =0.0001) when the plasmid encoding GM-CSF was coadministered with Sm-p80 DNA. Coinjection of plasmid DNA encoding IL-4 with Sm-p80 DNA yielded a protection level of 42% (P = < or =0.0001). Statistically, the protection conferred by including GM-CSF, but not IL-4, was significantly greater than that when only Sm-p80 was used. Sm-p80 DNA by itself elicited strong responses that include IgG2A and IgG2B antibody isotypes. The introduction of GM-CSF DNA with Sm-p80 DNA led to distinct increases in total IgG and IgG1 titers, whereas the coadministration of IL-4 DNA with Sm-p80 DNA resulted in a slight elevation of IgG1 and IgG3 titers and in some reduction of IgG2A and IgG2B titers. Our data again indicate that Sm-p80 can be an excellent candidate for a schistosomiasis vaccine.
Collapse
Affiliation(s)
- Afzal A Siddiqui
- Department of Internal Medicine, Texas Tech University Health Sciences Center and Veterans Affairs Health Care System, Amarillo, Texas 79106, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Siddiqui AA, Phillips T, Charest H, Podesta RB, Quinlin ML, Pinkston JR, Lloyd JD, Pompa J, Villalovos RM, Paz M. Enhancement of Sm-p80 (large subunit of calpain) induced protective immunity against Schistosoma mansoni through co-delivery of interleukin-2 and interleukin-12 in a DNA vaccine formulation. Vaccine 2003; 21:2882-9. [PMID: 12798631 DOI: 10.1016/s0264-410x(03)00159-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Schistosomiasis afflicts an estimated 200 million people in 76 countries and an additional 600 million people are at risk of acquiring this infection. Even though effective anthelmintic treatment and snail eradication control programs exist, the discovery of an effective vaccine still remains the most potentially powerful means of control for this disease. We have concentrated on a vaccine candidate (large subunit of calpain or Sm-p80) because of its potential in conferring protection against challenge infection and its pivotal role in surface membrane biogenesis of schistosomes. Since surface membrane renewal is a major phenomenon employed by hemohelminths to evade host immune system; an immune response directed against Sm-p80 should make the parasite prone to immune clearance from the host by both providing a well-targeted attack and by potentially inhibiting the surface membrane biogenesis process. In the present study, we have utilized DNA immunization protocols using Sm-p80 with plasmids encoding interleukin-2 (IL-2) and interleukin-12 (IL-12). Sm-p80 by itself provided a 39% protection (P</=0.0001) against challenge infection in C57BL/6 mice. This protection was increased to 57% (P</=0.0001) when plasmid encoding IL-2 was co-administered with Sm-p80 DNA. Co-injection of plasmid DNA encoding IL-12 with Sm-p80 DNA yielded a protection level of 45% (P</=0.0001). Statistically, the protection conferred by including IL-2 and IL-12 was significantly greater than when only the Sm-p80 was used. Sm-p80 DNA by itself elicited strong responses that includes IgG(2A) and IgG(2B) antibody isotypes. The introduction of IL-2 DNA with Sm-p80 DNA led to an increase in total IgG and IgG(2A) and IgG(2B) titres. Whereas co-administration of IL-12 DNA with Sm-p80 DNA resulted in the augmentation of only total IgG and IgG(2A). This data reinforces the potential of Sm-p80 as an excellent candidate for a schistosomiasis vaccine.
Collapse
Affiliation(s)
- Afzal A Siddiqui
- Department of Internal Medicine, Texas Tech Woman's Health & Research Institute, Texas Tech University Health Sciences Center, 1400 Wallace Blvd., Amarillo, Amarillo, TX 79106-1791, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Schistosomiasis continues to be a significant health problem, especially in Africa. While the recent World Health Assembly resolution to apply mass drug treatment of school age children in endemic areas is a very promising development, a preferable long-term solution would be provided by a protective vaccine. Encouragingly, one vaccine, Bilhvax, is in phase II trials. However, there is a danger that momentum towards the goal of developing a vaccine against schistosomiasis is dissipating. This article reviews the current situation in this important area of schistosomiasis research.
Collapse
Affiliation(s)
- Edward J Pearce
- Department of Pathobiology, Room 203D, Johnson Pavilion, 3610 Hamilton Walk, University of Pennsylvania, Philadelphia, PA 19104-6076, USA.
| |
Collapse
|
46
|
Shi F, Zhang Y, Lin J, Zuo X, Shen W, Cai Y, Ye P, Bickle QD, Taylor MG. Field testing of Schistosoma japonicum DNA vaccines in cattle in China. Vaccine 2002; 20:3629-31. [PMID: 12399187 DOI: 10.1016/s0264-410x(02)00398-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Vaccines are needed to reduce the zoonotic reservoir of Schistosoma japonicum infection in bovines in China. We have developed two experimental DNA vaccines and have already shown these to be capable of inducing partial protection in water buffalo naturally exposed to the risk of S. japonicum infection in the field. We now report a similar field trial in cattle, the other major bovine reservoir host species in China. Groups of cattle were vaccinated with the VRSj28 vaccine or the VRSj23 vaccine, or, to test whether protection could be enhanced by combination vaccination, with both these DNA vaccines together. After vaccination, the cattle were exposed to natural infection in the field for a period of 54 days. Worm and egg counts carried out at the end of the experiment showed that each of the vaccine groups showed partial resistance, and that combined vaccination was not more effective than vaccination with the individual plasmids.
Collapse
Affiliation(s)
- Fuhui Shi
- Shanghai Institute of Animal Parasitology, Chinese Academy of Agricultural Sciences, 3 Lane 345 Shi-long Road, Shanghai 200232, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|