1
|
Skelly PJ, Da'dara AA. A novel, non-neuronal acetylcholinesterase of schistosome parasites is essential for definitive host infection. Front Immunol 2023; 14:1056469. [PMID: 36798133 PMCID: PMC9927205 DOI: 10.3389/fimmu.2023.1056469] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 01/16/2023] [Indexed: 02/03/2023] Open
Abstract
Schistosomes are long-lived parasitic worms that infect >200 million people globally. The intravascular life stages are known to display acetylcholinesterase (AChE) activity internally as well as, somewhat surprisingly, on external tegumental membranes. Originally it was hypothesized that a single gene (SmAChE1 in Schistosoma mansoni) encoded both forms of the enzyme. Here, we demonstrate that a second gene, designated "S. mansoni tegumental acetylcholinesterase, SmTAChE", is responsible for surface, non-neuronal AChE activity. The SmTAChE protein is GPI-anchored and contains all essential amino acids necessary for function. AChE surface activity is significantly diminished following SmTAChE gene suppression using RNAi, but not following SmAChE1 gene suppression. Suppressing SmTAChE significantly impairs the ability of parasites to establish infection in mice, showing that SmTAChE performs an essential function for the worms in vivo. Living S. haematobium and S. japonicum parasites also display strong surface AChE activity, and we have cloned SmTAChE homologs from these two species. This work helps to clarify longstanding confusion regarding schistosome AChEs and paves the way for novel therapeutics for schistosomiasis.
Collapse
Affiliation(s)
- Patrick J Skelly
- Molecular Helminthology Laboratory, Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, United States
| | - Akram A Da'dara
- Molecular Helminthology Laboratory, Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, United States
| |
Collapse
|
2
|
Al-Naseri A, Al-Absi S, El Ridi R, Mahana N. A comprehensive and critical overview of schistosomiasis vaccine candidates. J Parasit Dis 2021; 45:557-580. [PMID: 33935395 PMCID: PMC8068781 DOI: 10.1007/s12639-021-01387-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 03/31/2021] [Indexed: 12/11/2022] Open
Abstract
A digenetic platyhelminth Schistosoma is the causative agent of schistosomiasis, one of the neglected tropical diseases that affect humans and animals in numerous countries in the Middle East, sub-Saharan Africa, South America and China. Several control methods were used for prevention of infection or treatment of acute and chronic disease. Mass drug administration led to reduction in heavy-intensity infections and morbidity, but failed to decrease schistosomiasis prevalence and eliminate transmission, indicating the need to develop anti-schistosome vaccine to prevent infection and parasite transmission. This review summarizes the efficacy and protective capacity of available schistosomiasis vaccine candidates with some insights and future prospects.
Collapse
Affiliation(s)
- Aya Al-Naseri
- Zoology Department, Faculty of Science, Cairo Univesity, Giza, 12613 Egypt
| | - Samar Al-Absi
- Zoology Department, Faculty of Science, Cairo Univesity, Giza, 12613 Egypt
| | - Rashika El Ridi
- Zoology Department, Faculty of Science, Cairo Univesity, Giza, 12613 Egypt
| | - Noha Mahana
- Zoology Department, Faculty of Science, Cairo Univesity, Giza, 12613 Egypt
| |
Collapse
|
3
|
Phumrattanaprapin W, Chaiyadet S, Brindley PJ, Pearson M, Smout MJ, Loukas A, Laha T. Orally Administered Bacillus Spores Expressing an Extracellular Vesicle-Derived Tetraspanin Protect Hamsters Against Challenge Infection With Carcinogenic Human Liver Fluke. J Infect Dis 2021; 223:1445-1455. [PMID: 32813017 PMCID: PMC8064041 DOI: 10.1093/infdis/jiaa516] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/10/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The human liver fluke Opisthorchis viverrini is a food-borne trematode that causes hepatobiliary disease in humans throughout Southeast Asia. People become infected by consuming raw or undercooked fish containing metacercariae. Development of a vaccine to prevent or minimize pathology would decrease the risk of severe morbidity, including the development of bile duct cancer. METHODS We produced an oral vaccine based on recombinant Bacillus subtilis spores expressing the large extracellular loop (LEL) of O. viverrini tetraspanin-2 (Ov-TSP-2), a protein that is abundant on the surface of O. viverrini secreted extracellular vesicles (EVs). Recombinant spores expressing Ov-TSP-2-LEL were orally administered to hamsters prior to challenge infection with O. viverrini metacercariae. RESULTS Vaccinated hamsters generated serum IgG as well as bile IgG and IgA responses to Ov-TSP-2-LEL, and serum IgG from vaccinated hamsters blocked the uptake of fluke EVs by a human bile duct epithelial cell line. Vaccinated hamsters had 56% reductions in both adult flukes and fecal eggs compared to the control group. CONCLUSIONS These findings indicate that oral vaccination of hamsters with recombinant B. subtilis spores expressing Ov-TSP-2-LEL is efficacious at reducing infection intensity and could form the basis of a vaccine for control of carcinogenic liver fluke infection in humans.
Collapse
Affiliation(s)
| | - Sujittra Chaiyadet
- Tropical Medicine Graduate Program, Academic Affairs, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Paul J Brindley
- Department of Microbiology, Immunology and Tropical Medicine, Research Center for Neglected Diseases of Poverty, School of Medicine and Health Sciences, George Washington University, Washington, District of Columbia, USA
| | - Mark Pearson
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Michael J Smout
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Alex Loukas
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Thewarach Laha
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
4
|
Mekonnen GG, Tedla BA, Pickering D, Becker L, Wang L, Zhan B, Bottazzi ME, Loukas A, Sotillo J, Pearson MS. Schistosoma haematobium Extracellular Vesicle Proteins Confer Protection in a Heterologous Model of Schistosomiasis. Vaccines (Basel) 2020; 8:E416. [PMID: 32722279 PMCID: PMC7563238 DOI: 10.3390/vaccines8030416] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/19/2020] [Accepted: 07/22/2020] [Indexed: 01/16/2023] Open
Abstract
Helminth parasites release extracellular vesicles which interact with the surrounding host tissues, mediating host-parasite communication and other fundamental processes of parasitism. As such, vesicle proteins present attractive targets for the development of novel intervention strategies to control these parasites and the diseases they cause. Herein, we describe the first proteomic analysis by LC-MS/MS of two types of extracellular vesicles (exosome-like, 120 k pellet vesicles and microvesicle-like, 15 k pellet vesicles) from adult Schistosoma haematobium worms. A total of 57 and 330 proteins were identified in the 120 k pellet vesicles and larger 15 k pellet vesicles, respectively, and some of the most abundant molecules included homologues of known helminth vaccine and diagnostic candidates such as Sm-TSP2, Sm23, glutathione S-transferase, saponins and aminopeptidases. Tetraspanins were highly represented in the analysis and found in both vesicle types. Vaccination of mice with recombinant versions of three of these tetraspanins induced protection in a heterologous challenge (S. mansoni) model of infection, resulting in significant reductions (averaged across two independent trials) in liver (47%, 38% and 41%) and intestinal (47%, 45% and 41%) egg burdens. These findings offer insight into the mechanisms by which anti-tetraspanin antibodies confer protection and highlight the potential that extracellular vesicle surface proteins offer as anti-helminth vaccines.
Collapse
Affiliation(s)
- Gebeyaw G. Mekonnen
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns 4878, Queensland, Australia; (G.G.M.); (B.A.T.); (D.P.); (L.B.); (J.S.)
- Department of Medical Parasitology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Bemnet A. Tedla
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns 4878, Queensland, Australia; (G.G.M.); (B.A.T.); (D.P.); (L.B.); (J.S.)
| | - Darren Pickering
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns 4878, Queensland, Australia; (G.G.M.); (B.A.T.); (D.P.); (L.B.); (J.S.)
| | - Luke Becker
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns 4878, Queensland, Australia; (G.G.M.); (B.A.T.); (D.P.); (L.B.); (J.S.)
| | - Lei Wang
- Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics and National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA; (L.W.); (B.Z.); (M.E.B.)
| | - Bin Zhan
- Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics and National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA; (L.W.); (B.Z.); (M.E.B.)
| | - Maria Elena Bottazzi
- Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics and National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA; (L.W.); (B.Z.); (M.E.B.)
| | - Alex Loukas
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns 4878, Queensland, Australia; (G.G.M.); (B.A.T.); (D.P.); (L.B.); (J.S.)
| | - Javier Sotillo
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns 4878, Queensland, Australia; (G.G.M.); (B.A.T.); (D.P.); (L.B.); (J.S.)
- Parasitology Reference and Research Laboratory, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain
| | - Mark S. Pearson
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns 4878, Queensland, Australia; (G.G.M.); (B.A.T.); (D.P.); (L.B.); (J.S.)
| |
Collapse
|
5
|
Da'Dara AA, Li C, Yu X, Zheng M, Zhou J, Shollenberger LM, Li YS, Harn DA. Prime-Boost Vaccine Regimen for SjTPI and SjC23 Schistosome Vaccines, Increases Efficacy in Water Buffalo in a Field Trial in China. Front Immunol 2019; 10:284. [PMID: 30842779 PMCID: PMC6391362 DOI: 10.3389/fimmu.2019.00284] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 02/04/2019] [Indexed: 01/14/2023] Open
Abstract
Schistosomiasis remains a serious zoonotic disease in China and the Philippines. Water buffalo and cattle account for the majority of transmission. Vaccination of water buffalo is considered a key strategy to reduce disease prevalence. Previously, we showed that vaccination of water buffalo with SjC23 or SjCTPI plasmid DNA vaccines, induced 50% efficacy to challenge infection. Here, we evaluated several parameters to determine if we can develop a two dose vaccine that maintains the efficacy of the three dose vaccine. We performed four trials evaluating: (1) lab produced vs. GLP grade vaccines, (2) varying the time between prime and boost, (3) the influence of an IL-12 adjuvant, and (4) a two dose heterologous (DNA-protein) prime-boost. We found the source of the DNA vaccines did not matter, nor did increasing the interval between prime and boost. Elimination of the IL-12 plasmid lowered homologous DNA-DNA vaccine efficacy. A major finding was that the heterologous prime boost improved vaccine efficacy, with the prime-boost regimen incorporating both antigens providing a 55% reduction in adult worms and 53% reduction in liver eggs. Vaccinated buffalo produced vaccine-specific antibody responses. These trials suggest that highly effective vaccination against schistosomes can be achieved using a two dose regimen. No adjuvants were used with the protein boost, and the potential that addition of adjuvant to the protein boost to further increase efficacy should be evaluated. These results suggest that use of these two schistosome vaccines can be part of an integrated control strategy to reduce transmission of schistosomiasis in Asia.
Collapse
Affiliation(s)
- Akram A. Da'Dara
- Department of Infectious Diseases and Global Health, Tufts Cummings School of Veterinary Medicine, Tufts University, Grafton, MA, United States
| | - Changlin Li
- Department of Infectious Diseases and Center for Tropical and Emerging Global Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Xinling Yu
- Hunan Institute of Parasitic Diseases, World Health Organisation Collaborating Centre for Research and Control of Schistosomiasis in Lake Region, Yueyang, China
| | - Mao Zheng
- Hunan Institute of Parasitic Diseases, World Health Organisation Collaborating Centre for Research and Control of Schistosomiasis in Lake Region, Yueyang, China
| | - Jie Zhou
- Hunan Institute of Parasitic Diseases, World Health Organisation Collaborating Centre for Research and Control of Schistosomiasis in Lake Region, Yueyang, China
| | - Lisa M. Shollenberger
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States
| | - Yue-sheng Li
- Hunan Institute of Parasitic Diseases, World Health Organisation Collaborating Centre for Research and Control of Schistosomiasis in Lake Region, Yueyang, China
- Molecular Parasitology Laboratory, Infectious Diseases Division, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Donald A. Harn
- Department of Infectious Diseases and Center for Tropical and Emerging Global Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| |
Collapse
|
6
|
White Bear J, Long T, Skinner D, McKerrow JH. Predictions of novel Schistosoma mansoni - human protein interactions consistent with experimental data. Sci Rep 2018; 8:13092. [PMID: 30166569 PMCID: PMC6117258 DOI: 10.1038/s41598-018-31272-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 08/14/2018] [Indexed: 12/26/2022] Open
Abstract
Infection by the human blood fluke, Schistosoma mansoni involves a variety of cross-species protein- protein interactions. The pathogen expresses a diverse arsenal of proteins that facilitate the breach of physical and biochemical barriers present in skin evasion of the immune system, and digestion of human plasma proteins including albumin and hemoglobin, allowing schistosomes to reside in the host for years. However, only a small number of specific interactions between S. mansoni and human proteins have been identified. We present and apply a protocol that generates testable predictions of S. mansoni-human protein interactions. In this study, we have preliminary predictions of novel interactions between schistosome and human proteins relevant to infection and the ability of the parasite to evade the immune system. We applied a computational whole-genome comparative approach to predict potential S. mansoni-human protein interactions based on similarity to known protein complexes. We first predict S. mansoni -human protein interactions based on similarity to known protein complexes. Putative interactions were then scored and assessed using several contextual filters, including the use of annotation automatically derived from literature using a simple natural language processing methodology. Next, in vitro experiments were carried out between schistosome and host proteins to validate several prospective predictions. Our method predicted 7 out of the 10 previously known cross-species interactions involved in pathogenesis between S. mansoni and its human host. Interestingly, two novel putative interactions involving Schistosoma proteins, the cercarial elastase SmCE, and the adult tegument surface protein Sm29, were also predicted and experimentally characterized. Preliminary data suggest that elafin, a host endogenous serine protease inhibitor, may be a novel substrate for SmCE. Additionally, CD59, an inhibitor of the membrane attack complex, could interact with Sm29. Furthermore, the application framework provides an integrated methodology for investigation of host-pathogen interactions and an extensive source of orthogonal data for experimental analysis. We have made the predictions available for community perusal.
Collapse
Affiliation(s)
- J White Bear
- Department of Bioengineering and Therapeutic Sciences, Department of Pharmaceutical Chemistry, and California Institute for Quantitative Biosciences, University of California, San Francisco, CA, 94158, USA.
- Graduate Group in Bioinformatics, University of California, San Francisco, CA, 94158, USA.
- MIT Lincoln Laboratory 244 Wood St, Lexington, MA, USA.
| | - Thavy Long
- Department of Pathology and Sandler Center for Basic Research in Parasitic Diseases, University of California at San Francisco, San Francisco, California, 94158, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego 9500 Gilman Dr, La Jolla, CA, 92093, USA
- INRA - InTheRes - UMR 1436, Equipe Transporteurs Membranaires et Résistance, 180, Chemin de Tournefeuille, Toulouse, France
| | - Danielle Skinner
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego 9500 Gilman Dr, La Jolla, CA, 92093, USA
| | - James H McKerrow
- Department of Pathology and Sandler Center for Basic Research in Parasitic Diseases, University of California at San Francisco, San Francisco, California, 94158, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego 9500 Gilman Dr, La Jolla, CA, 92093, USA
| |
Collapse
|
7
|
Gonçalves de Assis NR, Batistoni de Morais S, Figueiredo BCP, Ricci ND, de Almeida LA, da Silva Pinheiro C, Martins VDP, Oliveira SC. DNA Vaccine Encoding the Chimeric Form of Schistosoma mansoni Sm-TSP2 and Sm29 Confers Partial Protection against Challenge Infection. PLoS One 2015; 10:e0125075. [PMID: 25942636 PMCID: PMC4420270 DOI: 10.1371/journal.pone.0125075] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 03/19/2015] [Indexed: 12/23/2022] Open
Abstract
Schistosomiasis is an important parasitic disease worldwide that affects more than 207 million people in 76 countries and causes approximately 250,000 deaths per year. The best long-term strategy to control schistosomiasis is through immunization combined with drug treatment. Due to the ability of DNA vaccines to generate humoral and cellular immune responses, such vaccines are considered a promising approach against schistosomiasis. Sm29 and tetraspanin-2 (Sm-TSP2) are two proteins that are located in the S. mansoni tegument of adult worms and schistosomula and induce high levels of protection through recombinant protein immunization. In this study, we transfected BHK-21 cells with plasmids encoding Sm29, Sm-TSP2 or a chimera containing both genes. Using RT-PCR analysis and western blot, we confirmed that the DNA vaccine constructs were transcribed and translated, respectively, in BHK-21 cells. After immunization of mice, we evaluated the reduction in worm burden. We observed worm burden reductions of 17-22%, 22%, 31-32% and 24-32% in animals immunized with the pUMVC3/Sm29, pUMVC3/SmTSP-2, pUMVC3/Chimera and pUMVC3/Sm29 + pUMVC3/SmTSP-2 plasmids, respectively. We evaluated the humoral response elicited by DNA vaccines, and animals immunized with pUMVC3/Sm29 and pUMVC3/Sm29 + pUMVC3/SmTSP-2 showed higher titers of anti-Sm29 antibodies. The cytokine profile produced by the spleen cells of immunized mice was then evaluated. We observed higher production of Th1 cytokines, such as TNF-α and IFN-γ, in vaccinated mice and no significant production of IL-4 and IL-5. The DNA vaccines tested in this study showed the ability to generate a protective immune response against schistosomiasis, probably through the production of Th1 cytokines. However, future strategies aiming to optimize the protective response induced by a chimeric DNA construct need to be developed.
Collapse
Affiliation(s)
- Natan Raimundo Gonçalves de Assis
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270–901, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, 31270–901, Belo Horizonte, MG, Brazil
| | - Suellen Batistoni de Morais
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270–901, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, 31270–901, Belo Horizonte, MG, Brazil
| | - Bárbara Castro Pimentel Figueiredo
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270–901, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, 31270–901, Belo Horizonte, MG, Brazil
| | - Natasha Delaqua Ricci
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270–901, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, 31270–901, Belo Horizonte, MG, Brazil
| | - Leonardo Augusto de Almeida
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270–901, Belo Horizonte, MG, Brazil
| | - Carina da Silva Pinheiro
- Departamento de Biointeração do Instituto de Ciências da Saúde, Universidade Federal da Bahia, 40110–100, Salvador, BA, Brazil
| | | | - Sergio Costa Oliveira
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270–901, Belo Horizonte, MG, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, 31270–901, Belo Horizonte, MG, Brazil
- * E-mail:
| |
Collapse
|
8
|
Toet H, Piedrafita DM, Spithill TW. Liver fluke vaccines in ruminants: strategies, progress and future opportunities. Int J Parasitol 2014; 44:915-27. [PMID: 25200351 DOI: 10.1016/j.ijpara.2014.07.011] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 07/18/2014] [Accepted: 07/21/2014] [Indexed: 12/27/2022]
Abstract
The development of a vaccine for Fasciola spp. in livestock is a challenge and would be advanced by harnessing our knowledge of acquired immune mechanisms expressed by resistant livestock against fluke infection. Antibody-dependent cell-mediated cytotoxicity directed to the surface tegument of juvenile/immature flukes is a host immune effector mechanism, suggesting that antigens on the surface of young flukes may represent prime candidates for a fluke vaccine. A Type 1 immune response shortly after fluke infection is associated with resistance to infection in resistant sheep, indicating that vaccine formulations should attempt to induce Type 1 responses to enhance vaccine efficacy. In cattle or sheep, an optimal fluke vaccine would need to reduce mean fluke burdens in a herd below the threshold of 30-54 flukes to ensure sustainable production benefits. Fluke infection intensity data suggest that vaccine efficacy of approximately 80% is required to reduce fluke burdens below this threshold in most countries. With the increased global prevalence of triclabendazole-resistant Fasciolahepatica, it may be commercially feasible in the short term to introduce a fluke vaccine of reasonable efficacy that will provide economic benefits for producers in regions where chemical control of new drug-resistant fluke infections is not viable. Commercial partnerships will be needed to fast-track new candidate vaccines using acceptable adjuvants in relevant production animals, obviating the need to evaluate vaccine antigens in rodent models.
Collapse
Affiliation(s)
- Hayley Toet
- Department of Agricultural Sciences and Centre for AgriBioscience, La Trobe University, Bundoora, Victoria 3083, Australia
| | - David M Piedrafita
- School of Applied Sciences and Engineering, Federation University, Churchill, Victoria 3842, Australia
| | - Terry W Spithill
- Department of Agricultural Sciences and Centre for AgriBioscience, La Trobe University, Bundoora, Victoria 3083, Australia.
| |
Collapse
|
9
|
Bahgat MM. Interaction Between the Neglected Tropical Disease Human Schistosomiasis and HCV Infection in Egypt: a Puzzling Relationship. J Clin Transl Hepatol 2014; 2:134-9. [PMID: 26356794 PMCID: PMC4521266 DOI: 10.14218/jcth.2013.00028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Revised: 02/21/2014] [Accepted: 02/26/2014] [Indexed: 02/06/2023] Open
Abstract
Egypt has the highest prevalence of chronic hepatitis C virus (HCV) infection and seropositivity worldwide, and it has been proposed that this enhanced susceptibility to HCV is related to coinfection with schistosomiasis. Although currently, there are no studies regarding the actual prevalence of both human schistosomiasis and schistosomiasis/HCV coinfection evidences strongly support that eliminating human schistosomiasis from Egypt is necessary to reduce both HCV prevalence and liver pathology. The present review highlights the significant impact of the neglected tropical disease human schistosomiasis on both susceptibility of Egyptians to HCV coinfection, severity of the resulting liver pathology, and poor response to antiviral therapy. The immune evasion mechanisms exerted by the HCV-NS3/4A protease domain, and the possible impact of immune evasion mechanisms exerted by proteases of larval, worm and egg stages of the parasite Schistosoma on human susceptibility to HCV infection are discussed. In addition, schistosome immune evasion mechanisms may include immunosuppression that in turn prevents clearance of HCV viremia and leads to relapsing HCV infection and severe liver pathology. I propose the generation of a replicon system from the most prevailing genotype (HCV-4a) in Egypt and establishing its replication on hepatoplastoma or immune cells in presence of bilharzial antigens. Finally, the use of a humanized small animal model that can acquire both HCV and S. mansoni infections will be important to further understand in real time the impact of coinfection on both the immune system and liver pathology.
Collapse
Affiliation(s)
- Mahmoud M. Bahgat
- Immunology and Infectious Diseases Group, Therapeutic Chemistry Department, the Centre of Excellence for Advanced Sciences, the National Research Centre, Dokki, Cairo, Egypt
- Research Group of Biomarkers for Infection and Immunity, Institute of Experimental Infection Research, TWINCORE Centre for Experimental and Clinical Infection Research, Hannover, Germany
| |
Collapse
|
10
|
Espíndola MS, Frantz FG, Soares LS, Masson AP, Tefé-Silva C, Bitencourt CS, Oliveira SC, Rodrigues V, Ramos SG, Silva CL, Faccioli LH. Combined immunization using DNA-Sm14 and DNA-Hsp65 increases CD8+ memory T cells, reduces chronic pathology and decreases egg viability during Schistosoma mansoni infection. BMC Infect Dis 2014; 14:263. [PMID: 24886395 PMCID: PMC4031977 DOI: 10.1186/1471-2334-14-263] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 05/07/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Schistosomiasis is one of the most important neglected diseases found in developing countries and affects 249 million people worldwide. The development of an efficient vaccination strategy is essential for the control of this disease. Previous work showed partial protection induced by DNA-Sm14 against Schistosoma mansoni infection, whereas DNA-Hsp65 showed immunostimulatory properties against infectious diseases, autoimmune diseases, cancer and antifibrotic properties in an egg-induced granuloma model. METHODS C57BL/6 mice received 4 doses of DNA-Sm14 (100 μg/dose) and DNA-Hsp65 (100 μg/dose), simultaneously administrated, or DNA-Sm14 alone, once a week, during four weeks. Three groups were included: 1- Control (no immunization); 2- DNA-Sm14; 3- DNA-Sm14/DNA-Hsp65. Two weeks following last immunization, animals were challenged subcutaneously with 30 cercariae. Fifteen, 48 and 69 days after infection splenocytes were collected to evaluate the number of CD8+ memory T cells (CD44(high)CD62(low)) using flow cytometry. Forty-eight days after challenge adult worms were collected by portal veins perfusion and intestines were collected to analyze the intestinal egg viability. Histological, immunohistochemical and soluble quantification of collagen and α-SMA accumulation were performed on the liver. RESULTS In the current work, we tested a new vaccination strategy using DNA-Sm14 with DNA-Hsp65 to potentiate the protection against schistosomiasis. Combined vaccination increased the number of CD8+ memory T cells and decreased egg viability on the intestinal wall of infected mice. In addition, simultaneous vaccination with DNA-Sm14/DNA-Hsp65 reduced collagen and α-SMA accumulation during the chronic phase of granuloma formation. CONCLUSION Simultaneous vaccination with DNA-Sm14/DNA-Hsp65 showed an immunostimulatory potential and antifibrotic property that is associated with the reduction of tissue damage on Schistosoma mansoni experimental infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Lúcia Helena Faccioli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av, do Café s/n 14040-903 Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
11
|
Abstract
Parasitic diseases caused by protozoan and helminth parasites are among the leading causes of morbidity and mortality in tropical and subtropical regions of the world. Unfortunately, at present, there is no vaccine against any human parasitic disease. Conventional vaccine methods have largely failed against parasitic infections. This is due, in part, to the complexity of the parasite life cycle, the ability of the parasite to evade the immune system, and difficulties in identifying and eliciting the desired protective immune responses. The discovery of DNA vaccines has renewed hope for vaccine development against parasites. In the last decade, DNA vaccines were successful in inducing at least partial protection against several parasitic diseases. This review discusses the latest developments in DNA vaccines against tropical parasitic diseases.
Collapse
Affiliation(s)
- Akram A Da'dara
- Department of Immunology and Infectious Disease, Harvard School of Public Health, 665 Huntington Ave, Boston, MA 02115, USA.
| | | |
Collapse
|
12
|
Dakshinamoorthy G, Munirathinam G, Stoicescu K, Reddy MV, Kalyanasundaram R. Large extracellular loop of tetraspanin as a potential vaccine candidate for filariasis. PLoS One 2013; 8:e77394. [PMID: 24146990 PMCID: PMC3795629 DOI: 10.1371/journal.pone.0077394] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 09/02/2013] [Indexed: 12/22/2022] Open
Abstract
Lymphatic filariasis affects nearly 120 million people worldwide and mass preventive chemotherapy is currently used as a strategy to control this infection. This has substantially reduced the incidence of the infection in several parts of the world. However, a prophylactic vaccine would be more effective in preventing future infections and will supplement the mass chemotherapy efforts. Unfortunately, there is no licensed vaccine available currently to prevent this infection. Molecules expressed on the surface of the parasite are potential candidates for vaccine development as they are exposed to the host immune system. In this study we show that the large extracellular loop of tetraspanin (TSP LEL), a protein expressed on the cuticle of Brugia malayi and Wuchereria bancrofti is a potential vaccine candidate. Our results showed that BmTSP LEL is expressed on the surface of B. malayi infective third stage larvae (L3) and sera from human subjects who are putatively immune to lymphatic filariasis carry high titer of IgG1 and IgG3 antibodies against BmTSP LEL and WbTSP LEL. We also showed that these antibodies in the sera of human subjects can participate in the killing of B. malayi L3 in an antibody dependent cell-mediated cytotoxicity mechanism. Vaccination trials in mice showed that close to 64% protection were achieved against challenge infections with B. malayi L3. Immunized animals showed high titer of anti-WbTSP LEL IgG1, IgG2a and IgG2b antibodies in the sera and IFN-γ secreting cells in the spleen. Onchocerca volvulus another filarial parasite also expresses TSP LEL. Cross-reactivity studies showed that IgG1 antibody in the sera of endemic normal subjects, recognize OvTSP LEL. Similarly, anti-OvTSP LEL antibodies in the sera of subjects who are immune to O. volvulus were also shown to cross-react with rWbTSP LEL and rBmTSP LEL. These findings thus suggested that rTSP LEL can be developed as a potential vaccine candidate against multiple filarial infections.
Collapse
Affiliation(s)
- Gajalakshmi Dakshinamoorthy
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, Illinois, United States of America
| | - Gnanasekar Munirathinam
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, Illinois, United States of America
| | - Kristen Stoicescu
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, Illinois, United States of America
| | - Maryada Venkatarami Reddy
- Department of Biochemistry, Mahatma Gandhi Institute of Medical Sciences, Sevagram, Maharashtra, India
| | - Ramaswamy Kalyanasundaram
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, Illinois, United States of America
- * E-mail:
| |
Collapse
|
13
|
Xiong Y, Zhang M, Hong Y, Wei M, Ai D, Meng P, Han Y, Fu Z, Shi Y, Yang J, Lin J. Characterization Analysis of Schistosoma japonicum Plasma Membrane Repair Relative Gene Myoferlin. PLoS One 2013; 8:e66396. [PMID: 23823740 PMCID: PMC3688920 DOI: 10.1371/journal.pone.0066396] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 05/06/2013] [Indexed: 12/25/2022] Open
Abstract
Myoferlin is a member of the ferlin family of proteins, which are involved in plasma membrane repair, and has been identified as one of the tegument proteins of Schistosoma japonicum. The tegument proteins are potential candidates for vaccines and new drug targets. In this study, myoferlin of S. japonicum (SjMF) was cloned, expressed and characterized, the potential of SjMF recombinant protein (rSjMF) as a vaccine candidate was evaluated, and the effect of praziquantel on SjMF was detected by Real-time PCR. Immunofluorescence showed that this protein was mainly distributed on the surface of worms at different stages. Sequence analysis revealed that the SjMF open reading frame was conserved at all stages of the S. japonicum life cycle. And SjMF transcription was upregulated in 42-day-old worms, and was significantly higher in female worms. Western blotting revealed that rSjMF showed strong immunogenicity. The cytokine profile and IgG isotype analysis demonstrated that rSjMF plus ISA206 immunization induced a mixed T helper (Th)1/Th2 response. Purified rSjMF emulsified with ISA206 adjuvant significantly reduced worm burden from 21.8% to 23.21% and liver egg number from42.58% to 28.35%. Besides, SjMF transcription was downregulated when worms were exposed to low-dose praziquantel (PZQ) and upregulated when PZQ was degraded, accompanied by recovery of damaged tegument. When worms were exposed to high-dose PZQ, SjMF transcription was downregulated all the time and the damaged tegument did not recover. These findings indicated that SjMF is a potential vaccine against S. japonicum and provides the basis for further investigations into the biological function of SjMF.
Collapse
Affiliation(s)
- Yanian Xiong
- National Laboratory of Animal Schistosomiasis Control/Key Laboratory of Animal Parasitology, Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
| | - Ming Zhang
- National Laboratory of Animal Schistosomiasis Control/Key Laboratory of Animal Parasitology, Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
| | - Yang Hong
- National Laboratory of Animal Schistosomiasis Control/Key Laboratory of Animal Parasitology, Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
| | - Meimei Wei
- National Laboratory of Animal Schistosomiasis Control/Key Laboratory of Animal Parasitology, Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
| | - Dezhou Ai
- National Laboratory of Animal Schistosomiasis Control/Key Laboratory of Animal Parasitology, Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
| | - Peipei Meng
- National Laboratory of Animal Schistosomiasis Control/Key Laboratory of Animal Parasitology, Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
| | - Yanhui Han
- National Laboratory of Animal Schistosomiasis Control/Key Laboratory of Animal Parasitology, Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
| | - Zhiqiang Fu
- National Laboratory of Animal Schistosomiasis Control/Key Laboratory of Animal Parasitology, Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
| | - Yaojun Shi
- National Laboratory of Animal Schistosomiasis Control/Key Laboratory of Animal Parasitology, Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
| | - Jianmei Yang
- National Laboratory of Animal Schistosomiasis Control/Key Laboratory of Animal Parasitology, Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
| | - Jiaojiao Lin
- National Laboratory of Animal Schistosomiasis Control/Key Laboratory of Animal Parasitology, Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, People’s Republic of China
| |
Collapse
|
14
|
Lochmatter C, Schneider CL, Ingram K, Keiser J, Schifferli JA. Schistosoma mansoni tetraspanning orphan receptor (SmTOR): a new vaccine candidate against schistosomiasis. Clin Exp Immunol 2013; 170:342-57. [PMID: 23121675 DOI: 10.1111/j.1365-2249.2012.04667.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
One approach to fight against schistosomiasis is to develop an efficient vaccine. Schistosoma mansoni tetraspanning orphan receptor (SmTOR) might be a vaccine candidate, as it is a tegument membrane protein expressed most highly in cercariae. In this study we characterized the recombinant first extracellular domain of SmTOR (rSmTORed1) as having the expected property to bind C2 of complement similarly to a smaller peptide of the same domain, and to produce specific and high-titre antibodies in BALB/c mice immunized using complete Freund's adjuvant/incomplete Freund's adjuvant (CFA/IFA). Immunization was protective against parasite infection, as demonstrated by a significant decrease in worm burden in immunized BALB/c mice versus the control groups over two independent trials [64 and 45% reduction for mean adult worm burden in immunized versus phosphate-bufferd saline (PBS) injected mice]. Interestingly, infection by itself did not lead to the generation of anti-rSmTORed1 antibodies, corresponding to the low frequency of specific anti-rSmTORed1 antibodies detected in the sera of patients infected with S. mansoni (2/20; 10%). These data suggest that, as opposed to the natural infection during which SmTOR induces antibodies only rarely, immunization with its smaller first extracellular domain might be more efficient.
Collapse
Affiliation(s)
- C Lochmatter
- Immunonephrology Laboratory, Department of Biomedicine, University Hospital Basel, Basel, Switzerland.
| | | | | | | | | |
Collapse
|
15
|
Ma Y, An HJ, Wei XQ, Xu Q, Yu YZ, Sun ZW. Enhanced potency of replicon vaccine using one vector to simultaneously co-express antigen and interleukin-4 molecular adjuvant. Hum Vaccin Immunother 2013; 9:242-9. [PMID: 23291932 DOI: 10.4161/hv.22888] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
We evaluated the utility of interleukin-4 (IL-4) as molecular adjuvant of replicon vaccines for botulinum neurotoxin serotype A (BoNT/A) in mouse model. In both Balb/c and C57/BL6 mice that received the plasmid DNA replicon vaccines derived from Semliki Forest virus (SFV) encoding the Hc gene of BoNT/A (AHc), the immunogenicity was significantly modulated and enhanced by co-delivery or co-express of the IL-4 molecular adjuvant. The enhanced potencies were also produced by co-delivery or co-expression of the IL-4 molecular adjuvant in mice immunized with the recombinant SFV replicon particles (VRP) vaccines. In particular, when AHc and IL-4 were co-expressed within the same replicon vaccine vector using dual-expression or bicistronic IRES, the anti-AHc antibody titers, serum neutralization titers and survival rates of immunized mice after challenged with BoNT/A were significantly increased. These results indicate IL-4 is an effective Th2-type adjuvant for the replicon vaccines in both strain mice, and the co-expression replicon vaccines described here may be an excellent candidate for further vaccine development in other animals or humans. Thus, we described a strategy to design and develop efficient vaccines against BoNT/A or other pathogens using one replicon vector to simultaneously co-express antigen and molecular adjuvant.
Collapse
Affiliation(s)
- Yao Ma
- College of Life Science and Bioengineering; Beijing Jiaotong University; Beijing, PR China
| | | | | | | | | | | |
Collapse
|
16
|
Gene gun bombardment with DNA-coated golden particles enhanced the protective effect of a DNA vaccine based on thioredoxin glutathione reductase of Schistosoma japonicum. BIOMED RESEARCH INTERNATIONAL 2012; 2013:952416. [PMID: 23509820 PMCID: PMC3591203 DOI: 10.1155/2013/952416] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 08/19/2012] [Accepted: 09/09/2012] [Indexed: 12/24/2022]
Abstract
Schistosomiasis, caused by infection with Schistosoma species, remains an important parasitic zoonosis. Thioredoxin glutathione reductase of Schistosoma japonicum (SjTGR) plays an important role in the development of the parasite and for its survival. Here we present a recombinant plasmid DNA vaccine, pVAX1/SjTGR, to estimate its protection against S. japonicum in BALB/c mice. The DNA vaccine administrated by particle bombardment induced higher protection than by intramuscular injection. All animals vaccinated with pVAX1/SjTGR developed significant specific anti-SjTGR antibodies than control groups. Moreover, animals immunized by gene gun exhibited a splenocyte proliferative response, with an increase in IFN-γ and IL-4. The recombinant plasmid administrated by gene gun achieved a medium protective efficacy of 27.83–38.83% (P < 0.01) of worm reduction and 40.38–44.51% (P < 0.01) of liver egg count reduction. It suggests that different modes of administering a DNA vaccine can influence the protective efficacy induced by the vaccine. Interestingly, from the enzymatic activity results, we found that worms obtained from pVAX1/SjTGR-vaccinated animals expressed lower enzymatic activity than the control group and the antibodies weakened the enzymatic activity of SjTGR in vitro, too. It implies that the high-level antibodies may contribute to the protective effects.
Collapse
|
17
|
Dang Z, Yagi K, Oku Y, Kouguchi H, Kajino K, Matsumoto J, Nakao R, Wakaguri H, Toyoda A, Yin H, Sugimoto C. A pilot study on developing mucosal vaccine against alveolar echinococcosis (AE) using recombinant tetraspanin 3: Vaccine efficacy and immunology. PLoS Negl Trop Dis 2012; 6:e1570. [PMID: 22479658 PMCID: PMC3313938 DOI: 10.1371/journal.pntd.0001570] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 02/05/2012] [Indexed: 12/13/2022] Open
Abstract
Background We have previously evaluated the vaccine efficacies of seven tetraspanins of Echinococcus multilocularis (Em-TSP1–7) against alveolar echinococcosis (AE) by subcutaneous (s.c.) administration with Freund's adjuvant. Over 85% of liver cyst lesion number reductions (CLNR) were achieved by recombinant Em-TSP1 (rEm-TSP1) and -TSP3 (rEm-TSP3). However, to develop an efficient and safe human vaccine, the efficacy of TSP mucosal vaccines must be thoroughly evaluated. Methodology/Principal Findings rEm-TSP1 and -TSP3 along with nontoxic CpG ODN (CpG oligodeoxynucleotides) adjuvant were intranasally (i.n.) immunized to BALB/c mice and their vaccine efficacies were evaluated by counting liver CLNR (experiment I). 37.1% (p<0.05) and 62.1% (p<0.001) of CLNR were achieved by these two proteins, respectively. To study the protection-associated immune responses induced by rEm-TSP3 via different immunization routes (i.n. administration with CpG or s.c. immunization with Freund's adjuvant), the systemic and mucosal antibody responses were detected by ELISA (experiment II). S.c. and i.n. administration of rEm-TSP3 achieved 81.9% (p<0.001) and 62.8% (p<0.01) CLNR in the liver, respectively. Both the immunization routes evoked strong serum IgG, IgG1 and IgG2α responses; i.n. immunization induced significantly higher IgA responses in nasal cavity and intestine compared with s.c. immunization (p<0.001). Both immunization routes induced extremely strong liver IgA antibody responses (p<0.001). The Th1 and Th2 cell responses were assessed by examining the IgG1/IgG2α ratio at two and three weeks post-immunization. S.c. immunization resulted in a reduction in the IgG1/IgG2α ratio (Th1 tendency), whereas i.n. immunization caused a shift from Th1 to Th2. Moreover, immunohistochemistry showed that Em-TSP1 and -TSP3 were extensively located on the surface of E. multilocularis cysts, protoscoleces and adult worms with additional expression of Em-TSP3 in the inner part of protoscoleces and oncospheres. Conclusions Our study indicated that i.n. administration of rEm-TSP3 with CpG is able to induce both systemic and local immune responses and thus provides significant protection against AE. Humans and rodents become infected with E. multilocularis by oral ingesting of the eggs, which then develop into cysts in the liver and progress an endless proliferation. Untreated AE has a fatality rate of >90% in humans. Tetraspanins have been identified in Schistosoma and showed potential as the prospective vaccine candidates. In our recent study, we first identified seven tetraspanins in E. multilocularis and evaluated their protective efficacies as vaccines against AE when subcutaneously administered to BALB/c mice. Mucosal immunization of protective proteins is able to induce strong local and systemic immune responses, which might play a crucial role in protecting humans against E. multilocularis infection via the intestine, blood and liver. We focused on Em-TSP3, which achieved significant vaccine efficacy via both s.c. and i.n. routes. The adjuvanticity of nontoxic CpG OND as i.n. vaccine adjuvant was evaluated. The widespread expression of Em-TSP3 in all the developmental stages of E. multilocularis, and the strong local and systemic immune responses evoked by i.n. administration of rEm-TSP3 with CpG OND adjuvant suggest that this study might open the way for developing efficient, nontoxic human mucosal vaccines against AE.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antibodies, Helminth/blood
- Antigens, Helminth/genetics
- Antigens, Helminth/immunology
- Echinococcosis
- Echinococcosis, Hepatic/prevention & control
- Echinococcus multilocularis/isolation & purification
- Enzyme-Linked Immunosorbent Assay
- Freund's Adjuvant/administration & dosage
- Glycoproteins/genetics
- Glycoproteins/immunology
- Immunity, Mucosal
- Immunoglobulin A/analysis
- Immunoglobulin G/blood
- Intestinal Mucosa/immunology
- Liver/parasitology
- Male
- Mice
- Mice, Inbred BALB C
- Nasal Mucosa/immunology
- Oligodeoxyribonucleotides/administration & dosage
- Pilot Projects
- Tetraspanins/genetics
- Tetraspanins/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Zhisheng Dang
- Division of Collaboration and Education, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Grazing Animal Diseases MOA, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Gansu, People's Republic of China
| | - Kinpei Yagi
- Department of Biological Science, Hokkaido Institute of Public Health, Sapporo, Hokkaido, Japan
| | - Yuzaburo Oku
- Parasitology Laboratory, School of Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Hirokazu Kouguchi
- Department of Biological Science, Hokkaido Institute of Public Health, Sapporo, Hokkaido, Japan
| | - Kiichi Kajino
- Division of Collaboration and Education, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Jun Matsumoto
- Laboratory of Medical Zoology, Nihon University College of Bioresource Sciences, Fujisawa, Japan
| | - Ryo Nakao
- Division of Collaboration and Education, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Hiroyuki Wakaguri
- Department of Medical Genome Science, Graduate School of Frontier Science, The University of Tokyo, Tokyo, Japan
| | - Atsushi Toyoda
- RIKEN Genomic Sciences Center, Yokohama, Kanagawa, Japan
- Comparative Genomics Laboratory, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Hong Yin
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Grazing Animal Diseases MOA, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Gansu, People's Republic of China
| | - Chihiro Sugimoto
- Division of Collaboration and Education, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
- * E-mail:
| |
Collapse
|
18
|
Pearson MS, Pickering DA, McSorley HJ, Bethony JM, Tribolet L, Dougall AM, Hotez PJ, Loukas A. Enhanced protective efficacy of a chimeric form of the schistosomiasis vaccine antigen Sm-TSP-2. PLoS Negl Trop Dis 2012; 6:e1564. [PMID: 22428079 PMCID: PMC3302818 DOI: 10.1371/journal.pntd.0001564] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 01/27/2012] [Indexed: 01/04/2023] Open
Abstract
The large extracellular loop of the Schistosoma mansoni tetraspanin, Sm-TSP-2, when fused to a thioredoxin partner and formulated with Freund's adjuvants, has been shown to be an efficacious vaccine against murine schistosomiasis. Moreover, Sm-TSP-2 is uniquely recognised by IgG1 and IgG3 from putatively resistant individuals resident in S. mansoni endemic areas in Brazil. In the present study, we expressed Sm-TSP-2 at high yield and in soluble form in E. coli without the need for a solubility enhancing fusion partner. We also expressed in E. coli a chimera called Sm-TSP-2/5B, which consisted of Sm-TSP-2 fused to the immunogenic 5B region of the hookworm aspartic protease and vaccine antigen, Na-APR-1. Sm-TSP-2 formulated with alum/CpG showed significant reductions in adult worm and liver egg burdens in two separate murine schistosomiasis challenge studies. Sm-TSP-2/5B afforded significantly greater protection than Sm-TSP-2 alone when both antigens were formulated with alum/CpG. The enhanced protection obtained with the chimeric fusion protein was associated with increased production of anti-Sm-TSP-2 antibodies and IL-4, IL-10 and IFN-γ from spleen cells of vaccinated animals. Sera from 666 individuals from Brazil who were infected with S. mansoni were screened for potentially deleterious IgE responses to Sm-TSP-2. Anti-Sm-TSP-2 IgE to this protein was not detected (also shown previously for Na-APR-1), suggesting that the chimeric antigen Sm-TSP-2/5B could be used to safely and effectively vaccinate people in areas where schistosomes and hookworms are endemic. There are currently no vaccines available to combat helminth (worm) infections in humans. The most devastating of the diseases caused by human helminths are schistosomiasis (or bilharzia) and hookworm disease. By fusing one of the lead schistosomiasis vaccine antigens, Sm-TSP-2, with a protective fragment from one of the lead hookworm vaccine antigens, Na-APR-1, we have produced a chimeric vaccine, termed Sm-TSP-2/5B that might provide protection against two debilitating and co-endemic neglected tropical diseases. Sm-TSP-2/5B provided increased protection compared to Sm-TSP-2 alone when formulated with human approved adjuvants and tested in a mouse model of schistosomiasis. Moreover, IgE against Sm-TSP-2 or Na-APR-1 has not been detected in the blood of residents from an area in Brazil that is endemic for schistosomes and hookworms, indicating that vaccines based on these molecules would be unlikely to generate allergic reactions in recipients from developing countries.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adolescent
- Adult
- Aged
- Alum Compounds/administration & dosage
- Animals
- Antibodies, Helminth/blood
- Antigens, Helminth/administration & dosage
- Antigens, Helminth/genetics
- Antigens, Helminth/immunology
- Aspartic Acid Proteases/administration & dosage
- Aspartic Acid Proteases/genetics
- Aspartic Acid Proteases/immunology
- Brazil
- Child
- Child, Preschool
- Cytokines/metabolism
- Disease Models, Animal
- Escherichia coli/genetics
- Female
- Gene Expression
- Humans
- Immunoglobulin G/blood
- Infant
- Leukocytes, Mononuclear/immunology
- Male
- Mice
- Mice, Inbred C57BL
- Middle Aged
- Oligodeoxyribonucleotides/administration & dosage
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Schistosomiasis/immunology
- Schistosomiasis/prevention & control
- Spleen/immunology
- Tetraspanins/administration & dosage
- Tetraspanins/genetics
- Tetraspanins/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/immunology
- Young Adult
Collapse
Affiliation(s)
- Mark S. Pearson
- Queensland Tropical Health Alliance and School of Public Health and Tropical Medicine, James Cook University, Cairns, Queensland, Australia
| | - Darren A. Pickering
- Queensland Tropical Health Alliance and School of Public Health and Tropical Medicine, James Cook University, Cairns, Queensland, Australia
| | - Henry J. McSorley
- Queensland Tropical Health Alliance and School of Public Health and Tropical Medicine, James Cook University, Cairns, Queensland, Australia
| | - Jeffrey M. Bethony
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University, Washington D.C., United States of America
| | - Leon Tribolet
- Queensland Tropical Health Alliance and School of Public Health and Tropical Medicine, James Cook University, Cairns, Queensland, Australia
| | - Annette M. Dougall
- Queensland Tropical Health Alliance and School of Public Health and Tropical Medicine, James Cook University, Cairns, Queensland, Australia
| | - Peter J. Hotez
- Department of Pediatrics and Molecular Virology and Microbiology, and National School of Tropical Medicine, Sabin Vaccine Institute and Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, Texas, United States of America
| | - Alex Loukas
- Queensland Tropical Health Alliance and School of Public Health and Tropical Medicine, James Cook University, Cairns, Queensland, Australia
- * E-mail:
| |
Collapse
|
19
|
Computational vaccinology: an important strategy to discover new potential S. mansoni vaccine candidates. J Biomed Biotechnol 2011; 2011:503068. [PMID: 22013383 PMCID: PMC3196198 DOI: 10.1155/2011/503068] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 08/12/2011] [Indexed: 11/17/2022] Open
Abstract
The flatworm Schistosoma mansoni is a blood fluke parasite that causes schistosomiasis, a debilitating disease that occurs throughout the developing world. Current schistosomiasis control strategies are mainly based on chemotherapy, but many researchers believe that the best long-term strategy to control schistosomiasis is through immunization with an antischistosomiasis vaccine combined with drug treatment. Several papers on Schistosoma mansoni vaccine and drug development have been published in the past few years, representing an important field of study. The advent of technologies that allow large-scale studies of genes and proteins had a remarkable impact on the screening of new and potential vaccine candidates in schistosomiasis. In this postgenomic scenario, bioinformatic technologies have emerged as important tools to mine transcriptomic, genomic, and proteomic databases. These new perspectives are leading to a new round of rational vaccine development. Herein, we discuss different strategies to identify potential S. mansoni vaccine candidates using computational vaccinology.
Collapse
|
20
|
Jiang Y, Xu X, Qing X, Pan W. Identification and characterization of six novel tetraspanins from Schistosoma japonicum. Parasit Vectors 2011; 4:190. [PMID: 21958506 PMCID: PMC3203850 DOI: 10.1186/1756-3305-4-190] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 09/29/2011] [Indexed: 11/18/2022] Open
Abstract
Background Tetraspanins (TSPs), also known as members of the trans-membrane 4 super-family (TM4SF), comprise an assemblage of surface antigens reported in eukaryotic organisms. In the work presented here, six novel TSP proteins from the human blood fluke Schistosoma japonicum (S. japonicum) were produced and analyzed through a combination of bioinformatics and experimental approaches. Results Six novel TSP proteins of Schistosoma japonicum (designated as Sj-TSP-#1~6) contained four trans-membrane regions and one large extracellular loop (LEL) with a conserved CCG motif. Size of the proteins varied from 227 to 291 amino acid residues. All the six proteins were produced in E.coli and immune sera to each protein were prepared. Analysis of transcription profiles of the proteins by RT-PCR showed that Sj-TSP-#4 was transcribed only in the egg stage while transcription of the Sj-TSP-#2 was detected in female worms but not in males. The similar results were obtained by Western blot. Immunolocalization of the TSP proteins by immunofluorescence assay showed that the Sj-TSP-#2, Sj-TSP-#5 and Sj-TSP-#6 were located in the tegument of worms. Conclusions This study provided six novel TSP members of S. japonicum including their sequences and recombinant proteins. Availability of the novel proteins and information on their expression profile and location provided a basis for further investigation of the TSP proteins for their biological functions and as vaccine candidates.
Collapse
Affiliation(s)
- Yanyan Jiang
- Institute for Infectious Diseases & Vaccine Development, Tongji University School of Medicine, 1239 Siping Road, Shanghai 200092, PR China
| | | | | | | |
Collapse
|
21
|
Castro-Borges W, Simpson DM, Dowle A, Curwen RS, Thomas-Oates J, Beynon RJ, Wilson RA. Abundance of tegument surface proteins in the human blood fluke Schistosoma mansoni determined by QconCAT proteomics. J Proteomics 2011; 74:1519-33. [PMID: 21704203 DOI: 10.1016/j.jprot.2011.06.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 06/06/2011] [Accepted: 06/12/2011] [Indexed: 12/20/2022]
Abstract
The schistosome tegument provides a major interface with the host blood stream in which it resides. Our recent proteomic studies have identified a range of proteins present in the complex tegument structure, and two models of protective immunity have implicated surface proteins as mediating antigens. We have used the QconCAT technique to evaluate the relative and absolute amounts of tegument proteins identified previously. A concatamer comprising R- or K-terminated peptides was generated with [(13)C(6)] lysine/arginine amino acids. Two tegument surface preparations were each spiked with the purified SmQconCAT as a standard, trypsin digested, and subjected to MALDI ToF-MS. The absolute amounts of protein in the biological samples were determined by comparing the areas under the pairs of peaks, separated by 6m/z units, representing the light and heavy peptides derived from the biological sample and SmQconCAT, respectively. We report that aquaporin is the most abundant transmembrane protein, followed by two phosphohydrolases. Tetraspanin Tsp-2 and Annexin-2 are also abundant but transporters are scarce. Sm200 surface protein comprised the bulk of the GPI-anchored fraction and likely resides in the secreted membranocalyx. Two host IgGs were identified but in amounts much lower than their targets. The findings are interpreted in relation to the models of protective immunity.
Collapse
Affiliation(s)
- William Castro-Borges
- Centre for Immunology & Infection, Department of Biology, University of York, Heslington, York, YO10 5DD, UK.
| | | | | | | | | | | | | |
Collapse
|
22
|
Wu C, Cai P, Chang Q, Hao L, Peng S, Sun X, Lu H, Yin J, Jiang N, Chen Q. Mapping the binding between the tetraspanin molecule (Sjc23) of Schistosoma japonicum and human non-immune IgG. PLoS One 2011; 6:e19112. [PMID: 21533061 PMCID: PMC3080413 DOI: 10.1371/journal.pone.0019112] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Accepted: 03/17/2011] [Indexed: 12/29/2022] Open
Abstract
Background Schistosomal parasites can establish parasitization in a human host for
decades; evasion of host immunorecognition including surface masking by
acquisition of host serum components is one of the strategies explored by
the parasites. Parasite molecules anchored on the membrane are the main
elements in the interaction. Sjc23, a member of the tetraspanin (TSP) family
of Schistosoma japonicum, was previously found to be highly
immunogenic and regarded as a vaccine candidate against schistosomiasis.
However, studies indicated that immunization with Sjc23 generated rapid
antibody responses which were less protective than that with other antigens.
The biological function of this membrane-anchored molecule has not been
defined after decades of vaccination studies. Methodology and Principal Findings In this study, we explored affinity pull-down and peptide competition assays
to investigate the potential binding between Sjc23 molecule and human
non-immune IgG. We determined that Sjc23 could bind human non-immune IgG and
the binding was through the interaction of the large extra-cellular domain
(LED) of Sjc23 (named Sjc23-LED) with the Fc domain of human IgG. Sjc23 had
no affinity to other immunoglobulin types. Affinity precipitation (pull-down
assay) in the presence of overlapping peptides further pinpointed to a
9-amino acid motif within Sjc23-LED that mediated the binding to human
IgG. Conclusion and Significance S. japonicum parasites cloak themselves through interaction
with human non-immune IgG, and a member of the tetraspanin family, Sjc23,
mediated the acquisition of human IgG via the interaction of a motif of 9
amino acids with the Fc domain of the IgG molecule. The consequence of this
interaction will likely benefit parasitism of S. japonicum
by evasion of host immune recognition or immunoresponses. This is the first
report that an epitope of schistosomal ligand and its immunoglobulin
receptor are defined, which provides further evidence of immune evasion
strategy adopted by S. japonicum.
Collapse
Affiliation(s)
- Chuang Wu
- Key Laboratory of Zoonosis, The Ministry of
Education, Jilin University, Changchun, China
| | - Pengfei Cai
- Laboratory of Parasitology, Institute of
Pathogen Biology/Institute of Basic Medicine, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing, China
| | - Qiaocheng Chang
- Key Laboratory of Zoonosis, The Ministry of
Education, Jilin University, Changchun, China
| | - Lili Hao
- College of Life Science and Technology,
Southwest University of Nationalities, Chengdu, China
| | - Shuai Peng
- Key Laboratory of Zoonosis, The Ministry of
Education, Jilin University, Changchun, China
| | - Xiaojing Sun
- Key Laboratory of Zoonosis, The Ministry of
Education, Jilin University, Changchun, China
| | - Huijun Lu
- Key Laboratory of Zoonosis, The Ministry of
Education, Jilin University, Changchun, China
| | - Jigang Yin
- Key Laboratory of Zoonosis, The Ministry of
Education, Jilin University, Changchun, China
| | - Ning Jiang
- Key Laboratory of Zoonosis, The Ministry of
Education, Jilin University, Changchun, China
- * E-mail: (NJ); (QC)
| | - Qijun Chen
- Key Laboratory of Zoonosis, The Ministry of
Education, Jilin University, Changchun, China
- Laboratory of Parasitology, Institute of
Pathogen Biology/Institute of Basic Medicine, Chinese Academy of Medical
Sciences and Peking Union Medical College, Beijing, China
- * E-mail: (NJ); (QC)
| |
Collapse
|
23
|
Zhu L, Liu HF, Lu MB, Long QK, Shi YE, Yu LJ. Construction, purification, and evaluation of multivalent DNA vaccine against Schistosoma japonicum. Parasitol Res 2010; 108:115-21. [PMID: 20852891 DOI: 10.1007/s00436-010-2040-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Accepted: 08/26/2010] [Indexed: 01/10/2023]
Abstract
DNA vaccine encoding a multivalent antigen is a novel approach of protective immunization. Four Schistosoma japonicum candidate antigen genes, glyceraldehyde-3-phosphate dehydrogenase (SjGAPDH), 23 kDa transmembrane protein (Sj23), 14 kDa fatty-acid binding protein (SjFABP) and 26 kDa glutathione-S-transferase (Sj26), are recombined into two pieces of fusion genes SjFABP.Sj23 and Sj26.SjGAPDH, respectively. Tetravalent DNA vaccine pVIVO2-SjFABP.Sj23/Sj26.SjGAPDH is constructed by co-expressing these two fusion genes. The super-coiled DNA vaccines for large-scale clinic application were purified by sequential chromatographies including group separation chromatography and affinitive chromatographies. The purified DNA vaccines were evaluated for in vivo and in vitro transfection assay. The immunoprotective properties of the different kinds of constructed DNA vaccines were appraised by pharmacological trials. The pharmacological trials results showed that tetravalent DNA vaccine has higher protective efficiency than other tested DNA vaccines.
Collapse
Affiliation(s)
- Lu Zhu
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China
| | | | | | | | | | | |
Collapse
|
24
|
Carvalho JA, Rodgers J, Atouguia J, Prazeres DMF, Monteiro GA. DNA vaccines: a rational design against parasitic diseases. Expert Rev Vaccines 2010; 9:175-91. [PMID: 20109028 DOI: 10.1586/erv.09.158] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Parasitic diseases are one of the most devastating causes of morbidity and mortality worldwide. Although immunization against these infections would be an ideal solution, the development of effective vaccines has been hampered by specific challenges posed by parasitic pathogens. Plasmid-based DNA vaccines may prove to be promising immunization tools in this area because vectors can be designed to integrate several antigens from different stages of the parasite life cycle or different subspecies; vaccines, formulations and immunization protocols can be tuned to match the immune response that offers protective immunity; and DNA vaccination is an affordable platform for developing countries. Partial and full protective immunity have been reported following DNA vaccination against the most significant parasitic diseases in the world.
Collapse
Affiliation(s)
- Joana A Carvalho
- Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | | | | | | | | |
Collapse
|
25
|
Mulvenna J, Moertel L, Jones MK, Nawaratna S, Lovas EM, Gobert GN, Colgrave M, Jones A, Loukas A, McManus DP. Exposed proteins of the Schistosoma japonicum tegument. Int J Parasitol 2009; 40:543-54. [PMID: 19853607 DOI: 10.1016/j.ijpara.2009.10.002] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Revised: 10/08/2009] [Accepted: 10/08/2009] [Indexed: 01/02/2023]
Abstract
The ability of the mammalian blood fluke Schistosoma japonicum to survive in the inhospitable environment of the mammalian bloodstream can be attributed, at least in part, to its host-exposed outer surface, called the tegument. The tegument is a dynamic organ and is involved in nutrition, immune evasion and modulation, excretion, osmoregulation and signal transduction. Given its importance for parasite survival, proteins exposed to the host at the surface of the tegument are ideal targets for the development of vaccines and drugs. By biotinylating live adult worms and using a combination of OFFGEL electrophoresis and tandem mass spectrometry 54 proteins were identified as putatively host-exposed in S. japonicum. These included glucose transport proteins, an amino permease, a leucine aminopeptidase and a range of transporters, heat shock proteins and novel immune-active proteins. Members of the tetraspanin protein family and a homologue of Sm 29, a tegument membrane protein from Schistosoma mansoni, both effective vaccine antigens in S. mansoni, were also identified. The fate of labelled surface proteins was monitored over time using electron microscopy and revealed that biotinylated proteins were rapidly internalised from the surface of the tegument and trafficked into the cytoplasmic bridges that connect the distal cytoplasm of the tegument to the underlying cell bodies. The results reported herein dramatically increase the number of S. japonicum proteins known to be exposed to the host and, hence, those of interest as therapeutic targets. The ability of the parasite to rapidly internalise proteins at its surface has implications for the development of vaccines and may explain how these parasites are able to avoid the host immune system for long periods of time.
Collapse
Affiliation(s)
- Jason Mulvenna
- Helminth Biology Laboratory, Division of Infectious Diseases, Queensland Institute of Medical Research, Qld 4006, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Han ZG, Brindley PJ, Wang SY, Chen Z. Schistosoma genomics: new perspectives on schistosome biology and host-parasite interaction. Annu Rev Genomics Hum Genet 2009; 10:211-40. [PMID: 19630560 DOI: 10.1146/annurev-genom-082908-150036] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Schistosomiasis, caused mainly by Schistosoma japonicum, S. mansoni, and S. hematobium, remains one of the most prevalent and serious parasitic diseases worldwide. The blood flukes have a complex life cycle requiring adaptation for survival in fresh water as free-living forms and as parasites in snail intermediate and vertebrate definitive hosts. Functional genomics analyses, including transcriptomic and proteomic approaches, have been performed on schistosomes, in particular S. mansoni and S. japonicum, using powerful high-throughput methodologies. These investigations have not only chartered gene expression profiles across genders and developmental stages within mammalian and snail hosts, but have also characterized the features of the surface tegument, the eggshell and excretory-secretory proteomes of schistosomes. The integration of the genomic, transcriptomic, and proteomic information, together with genetic manipulation on individual genes, will provide a global insight into the molecular architecture of the biology, pathogenesis, and host-parasite interactions of the human blood flukes. Importantly, these functional genomics analyses lay a foundation on which to develop new antischistosome vaccines as well as drug targets and diagnostic markers for treatment and control of schistosomiasis.
Collapse
Affiliation(s)
- Ze-Guang Han
- Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center at Shanghai, Shanghai 201203, China.
| | | | | | | |
Collapse
|
27
|
Evaluation of Echinococcus multilocularis tetraspanins as vaccine candidates against primary alveolar echinococcosis. Vaccine 2009; 27:7339-45. [PMID: 19782112 DOI: 10.1016/j.vaccine.2009.09.045] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Revised: 09/10/2009] [Accepted: 09/13/2009] [Indexed: 12/30/2022]
Abstract
Echinococcus multilocularis causes an important zoonotic cestode disease. The metacestode stage proliferates in the liver of intermediate hosts including human and rodents and forms multiple cysts. Recently, members of a transmembrane protein tetraspanin (TSP) family have been used as vaccines against schistosomosis, or as diagnostic antigens for cysticercosis. In this study, seven tetraspanins of E. multilocularis, designated as TSP1 to TSP7, were evaluated for their protective potential against primary alveolar echinococcosis. The large extracellular loop (LEL) region of these tetraspanins was cloned from a full-length enriched cDNA library of E. multilocularis metacestodes and expressed in Escherichia coli as a fusion protein with thioredoxin. Recombinant TSPs were applied as vaccines against an E. multilocularis primary experimental infection in BALB/c mice. Cyst lesions in the livers of vaccinated and non-vaccinated mice were counted. The cyst lesion reduction rates induced by the seven tetraspanins in vaccinated vis-à-vis non-vaccinated mice were: 87.9%, 65.8%, 85.1%, 66.9%, 73.7%, 72.9% and 37.6%. Vaccination conferred protective rates to mice ranging from 0% (TSP5, 6, 7) to maximally 33% (TSP1, 3). The results indicated that recombinant tetraspanins have varying protective effects against primary alveolar echinococcosis and could be used in vaccine development.
Collapse
|
28
|
Transforming growth factor-beta and Th17 responses in resistance to primary murine schistosomiasis mansoni. Cytokine 2009; 48:239-45. [PMID: 19717308 DOI: 10.1016/j.cyto.2009.07.581] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Revised: 07/23/2009] [Accepted: 07/31/2009] [Indexed: 01/05/2023]
Abstract
Discovery of the T-helper (Th) 17 cell lineage and functions in immune responses of mouse and man prompted us to investigate the role of transforming growth factor-beta (TGF-beta) and interleukin (IL)-17 in innate resistance to murine schistosomiasis mansoni. Schistosoma mansoni-infected BALB/c and C57BL/6 mice were administered with recombinant TGF-beta or mouse monoclonal antibody to TGF-beta to evaluate the impact of this cytokine on host immune responses against lung-stage schistosomula, and subsequent effects on adult worm parameters. Developing schistosomula elicited increase in peripheral blood mononuclear cells (PBMC) mRNA expression and/or plasma levels of IL-4, IL-17, and interferon-gamma (IFN-gamma), cytokines known to antagonize each other, resulting in impaired Th1/Th2, and Th17 immune responses and parasite evasion. Mice treated with TGF-beta showed elevated PBMC mRNA expression of IL-6, IL-17, TGF-beta, and TNF-alpha mRNA and increased IL-23 and IL-17 or TGF-beta plasma levels, associated with significantly (P<0.02-<0.0001) lower S. mansoni adult worm burden compared to controls in both mouse strains, thus suggesting that TGF-beta led to heightened Th17 responses that mediated resistance to the infection. Mice treated with antibody to TGF-beta showed increase in PBMC mRNA expression and plasma levels of IL-4, IL-12p70, and IFN-gamma, and significantly (P<0.02 and <0.0001) reduced worm burden and liver worm egg counts than untreated mice, indicating that Th1/Th2 immune responses were potentiated, resulting in significant innate resistance to schistosomiasis. The implications of these observations for schistosome immune evasion and vaccination were discussed.
Collapse
|
29
|
Oliveira SC, Fonseca CT, Cardoso FC, Farias LP, Leite LC. Recent advances in vaccine research against schistosomiasis in Brazil. Acta Trop 2008; 108:256-62. [PMID: 18577363 DOI: 10.1016/j.actatropica.2008.05.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Revised: 04/09/2008] [Accepted: 05/29/2008] [Indexed: 01/09/2023]
Abstract
Schistosomiasis continues to be a significant public health problem in tropical countries such as Brazil. Even though drug treatment in endemic areas has been shown to be efficient for controlling morbidity, it does not reduce prevalence due to constant reinfections. Therefore, a long-term disease control strategy is needed combining mass chemotherapy with a protective vaccine. Although the field of vaccine development has experienced more failures than successes, encouraging results have been obtained in recent years using defined recombinant derived Schistosoma mansoni antigens. This article primarily reviews the progress in the development of a vaccine against S. mansoni in Brazil. We discuss here different forms of vaccine tested in Brazil in pre-clinical trials and immunologic studies performed with patients in endemic areas of schistosomiasis. Lastly, we reviewed the S. mansoni genomic projects developed in the country and the recent advances in the identification of new molecules with potential as vaccine targets.
Collapse
|
30
|
Cardoso FC, Macedo GC, Gava E, Kitten GT, Mati VL, de Melo AL, Caliari MV, Almeida GT, Venancio TM, Verjovski-Almeida S, Oliveira SC. Schistosoma mansoni tegument protein Sm29 is able to induce a Th1-type of immune response and protection against parasite infection. PLoS Negl Trop Dis 2008; 2:e308. [PMID: 18827884 PMCID: PMC2553283 DOI: 10.1371/journal.pntd.0000308] [Citation(s) in RCA: 142] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2008] [Accepted: 09/04/2008] [Indexed: 01/08/2023] Open
Abstract
Background Schistosomiasis continues to be a significant public health problem. This disease affects 200 million people worldwide and almost 800 million people are at risk of acquiring the infection. Although vaccine development against this disease has experienced more failures than successes, encouraging results have recently been obtained using membrane-spanning protein antigens from the tegument of Schistosoma mansoni. Our group recently identified Sm29, another antigen that is present at the adult worm tegument surface. In this study, we investigated murine cellular immune responses to recombinant (r) Sm29 and tested this protein as a vaccine candidate. Methods and Findings We first show that Sm29 is located on the surface of adult worms and lung-stage schistosomula through confocal microscopy. Next, immunization of mice with rSm29 engendered 51%, 60% and 50% reduction in adult worm burdens, in intestinal eggs and in liver granuloma counts, respectively (p<0.05). Protective immunity in mice was associated with high titers of specific anti-Sm29 IgG1 and IgG2a and elevated production of IFN-γ, TNF-α and IL-12, a typical Th1 response. Gene expression analysis of worms recovered from rSm29 vaccinated mice relative to worms from control mice revealed a significant (q<0.01) down-regulation of 495 genes and up-regulation of only 22 genes. Among down-regulated genes, many of them encode surface antigens and proteins associated with immune signals, suggesting that under immune attack schistosomes reduce the expression of critical surface proteins. Conclusion This study demonstrates that Sm29 surface protein is a new vaccine candidate against schistosomiasis and suggests that Sm29 vaccination associated with other protective critical surface antigens is the next logical strategy for improving protection. Schistosomiasis is the most important human helminth infection in terms of morbidity and mortality. Although the efforts to develop a vaccine against this disease have experienced failures, a new generation of surface antigens revealed by proteomic studies changed this scenario. Our group has characterized the protein Sm29 described previously as one of the most exposed and expressed antigens in the outer tegument of Schistosoma mansoni. Studies in patients living in endemic areas for schistosomiasis revealed high levels of IgG1 and IgG3 anti-Sm29 in resistant individuals. In this study, confocal microscope analysis showed Sm29 present in the surface of lung-stage schistosoluma and adult worms. Recombinant Sm29, when used as vaccine candidate, induced high levels of protection in mice. This protection was associated with a typical Th1 immune response and reduction of worm burden, liver granulomas and in intestinal eggs. Further, microarray analysis of worms recovered from vaccinated mice showed significant down-regulation of several genes encoding previously characterized vaccine candidates and/or molecules exposed on the surface, suggesting an immune evasion strategy of schistosomes under immune attack. These results demonstrated that Sm29 as one of the important antigens with potential to compose a vaccine against schistosomiasis.
Collapse
Affiliation(s)
- Fernanda C. Cardoso
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gilson C. Macedo
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Elisandra Gava
- Departamento de Morfologia do Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gregory T. Kitten
- Departamento de Morfologia do Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vitor L. Mati
- Departamento de Parasitologia do Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Alan L. de Melo
- Departamento de Parasitologia do Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marcelo V. Caliari
- Departamento de Patologia do Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Giulliana T. Almeida
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Thiago M. Venancio
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | | | - Sergio C. Oliveira
- Departamento de Bioquímica e Imunologia do Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- * E-mail:
| |
Collapse
|
31
|
Da'dara AA, Li YS, Xiong T, Zhou J, Williams GM, McManus DP, Feng Z, Yu XL, Gray DJ, Harn DA. DNA-based vaccines protect against zoonotic schistosomiasis in water buffalo. Vaccine 2008; 26:3617-25. [PMID: 18524429 PMCID: PMC2567122 DOI: 10.1016/j.vaccine.2008.04.080] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Revised: 04/22/2008] [Accepted: 04/29/2008] [Indexed: 01/23/2023]
Abstract
Schistosomiasis japonica is an endemic, zoonotic disease of major public health importance in China where water buffaloes account for approximately 75% of disease transmission. Interventions that reduce schistosome infection in water buffaloes will enhance their health simultaneously reducing disease transmission to humans. While chemotherapy has proved successful, it requires continued time consuming and expensive mass treatments. A more sustainable option would be development of vaccines that reduce transmission of S. japonicum from bovines to replace bovine chemotherapy. We performed two randomized double blind trials in water buffaloes to determine if DNA vaccines encoding triose-phosphate isomerase (SjCTPI), or the tetraspanin 23 kDa integral membrane protein (SjC23), alone or fused to bovine heat shock protein 70 (Hsp70) could induce a level of immunity conducive to long-term sustainable control. Groups of water buffaloes (15/group) received three intramuscular injections, 4 weeks apart. Booster immunizations were co-administered with a plasmid DNA encoding IL-12. Four weeks after the last injection, water buffaloes were challenged with 1000 cercariae, and vaccine efficacy analyzed 8 weeks later. Water buffaloes vaccinated with SjCTPI-Hsp70 or SjCTPI plasmids had worm burdens reduced by 51.2% and 41.5%, respectively. Importantly, fecal miracidial hatching was reduced by 52.1% and 33.2% respectively compared to control vaccinated water buffaloes. Vaccination with SjC23-Hsp70 and SjC23 plasmids reduced worm burdens by 50.9% and 45.5%, respectively, and fecal miracidial hatching by 52.0% and 47.4%. A mathematical model of schistosome transmission predicts that schistosome vaccines capable of reducing water buffaloes' fecal egg output by 45%, alone or in conjunction with praziquantel treatment, will lead to a significant reduction in transmission of schistosomiasis. Both DNA vaccines tested here exceed this hypothetical level. Indeed, mathematical modeling of SjCTPI-Hsp70 and SjC23-Hsp70 alone and in conjunction with human chemotherapy showed a significant reduction in transmission almost to the point of elimination.
Collapse
Affiliation(s)
- Akram A Da'dara
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, 665 Huntington Avenue, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Bethony JM, Diemert DJ, Oliveira SC, Loukas A. Can schistosomiasis really be consigned to history without a vaccine? Vaccine 2008; 26:3373-6. [PMID: 18513839 DOI: 10.1016/j.vaccine.2008.04.045] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2007] [Revised: 03/17/2008] [Accepted: 04/03/2008] [Indexed: 12/27/2022]
Abstract
Recently, considerable enthusiasm has been expressed for expanding and combining control efforts for neglected tropical diseases (NTDs). While these efforts are laudable, the drugs in question require repeated mass administration for indefinite periods of time, and their use to achieve eradication is fraught with challenges. Mass drug administration is unlikely to be effective in isolation, and should not proceed without concurrent control methods, such as vaccines. Schistosomiasis is one of the most important NTDs, and one whose effective control is unlikely in the absence of improved sanitation and a vaccine. Recent advances in biotechnologies have enhanced antigen discovery and new molecules that show promise as recombinant vaccines are being reported. Funding bodies supporting research into the control of schistosomiasis should invest not only in mass drug administration but also in the development of new control strategies, including the development of vaccines.
Collapse
|
33
|
Romeih MH, Hassan HM, Shousha TSA, Saber MA. Immunization against Egyptian Schistosoma mansoni infection by multivalent DNA vaccine. Acta Biochim Biophys Sin (Shanghai) 2008; 40:327-38. [PMID: 18401531 DOI: 10.1111/j.1745-7270.2008.00404.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The development of multivalent vaccines consisting of several antigens is a novel approach to creating broad-range protection against different parasite strains and parasite life cycle stages. We have previously confirmed that the schistosome Sm21.7 and SmFimbrin (SmFim) proteins could induce protection in mice. Therefore, this study aimed to construct the multivalent DNA vaccine Sm21.7-SmFim/pBudCE4.1 and evaluate its immune efficacy. The open reading frames of two Schistosoma mansoni genes, Sm21.7 and SmFim, were inserted into the eukaryotic expression plasmid pBudCE4.1 designed for the independent expression of two genes in mammalian cells. To evaluate the in vitro expression of the multivalent Sm21.7-SmFim/pBudCE4.1 DNA vaccine and its immunological effect in mice, the recombinant plasmid Sm21.7-SmFim/pBudCE4.1 was used to transfect 293T cells, and the expression of mRNA and proteins was examined using reverse transcription-polymerase chain reaction and Western blot analysis. Then the ability of Sm21.7-SmFim/pBudCE4.1 to protect against S. mansoni challenge infections was analyzed according to worm burden and egg reduction rates after vaccination of mice. Vaccinated mice showed a significant level of protection (56%), and a decrease in the number and size, and change in the cellular profile, of granulomas. Egg reduction in liver and intestine was 41.53% and 55.63%, respectively, as determined relative to mice that received the empty vector only. In addition to reductions in worm viability, worm fecundity and egg hatching ability were observed following challenge infection in the immunized group. Results showed that Sm21.7-SmFim/pBudCE4.1 could express Sm21.7 and SmFim mRNA and proteins. Enzyme-linked immunosorbent assay and Western blot analysis indicated that immunized mice generated specific immunoglobulin G against Sm21.7-SmFim/pBudCE4.1. These results suggest that vaccination with multivalent S. mansoni DNA vaccine (SmFim-Sm21.7/pBudCE4.1) not only induces a significant reduction in worm and egg burdens, but also significantly reduces the size of egg granulomas. In summary, the multivalent vaccine stimulated specific immunity with a significant level of protection and has anti-pathological effect.
Collapse
Affiliation(s)
- Mahmoud H Romeih
- Department of Biochemistry and Molecular Biology, Theodor Bilharz Research Institute, Giza, Egypt.
| | | | | | | |
Collapse
|
34
|
Zhang L, Yang Y, Yang X, Zhao J, Yang J, Liu F, Zhang Z, Wu G, Su C. T cell epitope-based peptide-DNA dual vaccine induces protective immunity against Schistosoma japonicum infection in C57BL/6J mice. Microbes Infect 2008; 10:251-9. [DOI: 10.1016/j.micinf.2007.11.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2007] [Revised: 10/26/2007] [Accepted: 11/26/2007] [Indexed: 10/22/2022]
|
35
|
Abstract
Schistosomiasis, caused by trematode blood flukes of the genus Schistosoma, is recognized as the most important human helminth infection in terms of morbidity and mortality. Infection follows direct contact with freshwater harboring free-swimming larval (cercaria) forms of the parasite. Despite the existence of the highly effective antischistosome drug praziquantel (PZQ), schistosomiasis is spreading into new areas, and although it is the cornerstone of current control programs, PZQ chemotherapy does have limitations. In particular, mass treatment does not prevent reinfection. Furthermore, there is increasing concern about the development of parasite resistance to PZQ. Consequently, vaccine strategies represent an essential component for the future control of schistosomiasis as an adjunct to chemotherapy. An improved understanding of the immune response to schistosome infection, both in animal models and in humans, suggests that development of a vaccine may be possible. This review considers aspects of antischistosome protective immunity that are important in the context of vaccine development. The current status in the development of vaccines against the African (Schistosoma mansoni and S. haematobium) and Asian (S. japonicum) schistosomes is then discussed, as are new approaches that may improve the efficacy of available vaccines and aid in the identification of new targets for immune attack.
Collapse
|
36
|
Siddiqui AA, Ahmad G, Damian RT, Kennedy RC. Experimental vaccines in animal models for schistosomiasis. Parasitol Res 2008; 102:825-33. [PMID: 18259777 DOI: 10.1007/s00436-008-0887-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Accepted: 01/13/2008] [Indexed: 01/06/2023]
Abstract
Considerable morbidity and mortality results from the affliction of an estimated 200 million people worldwide by several species of schistosomes; 779 million are exposed to the disease in 74 different countries. Even though anti-parasitic drugs and other control measures, including public hygiene and snail control are available, the advent of an effective vaccine still remains the most potentially powerful means for the control of this disease. The putative vaccine could be administered to small children prior to the time when their contact with infected water is maximal, so as to prevent severe infection in the subsequent years. This review attempts to summarize the status of schistosome vaccine development with special emphasis on functionally important vaccine candidates. The importance of utilizing both murine and nonhuman primate models as a prerequisite for clinical trials is discussed.
Collapse
Affiliation(s)
- Afzal A Siddiqui
- Department of Microbiology and Immunology, Texas Tech University Health Sciences Center, 3601 4th Street, Stop 6591, Lubbock, TX 79430-6591, USA.
| | | | | | | |
Collapse
|
37
|
PEARSON MS, McMANUS DP, SMYTH DJ, JONES MK, SYKES AM, LOUKAS A. Cloning and characterization of an orphan seven transmembrane receptor from Schistosoma mansoni. Parasitology 2007; 134:2001-8. [PMID: 17714602 PMCID: PMC2753299 DOI: 10.1017/s0031182007003393] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A partial cDNA sequence was obtained from the human blood fluke, Schistosoma mansoni using a signal sequence trap approach. The full-length cDNA was cloned and termed Sm-7TM. The corresponding open reading frame had 7 membrane spanning domains and shared identity with a small, novel group of seven transmembrane (7TM) receptors from vertebrates and invertebrates, including the human ee3 receptor - a heptahelical protein implicated in neuronal signalling. Phylogenetic analysis of this novel family showed that the Sm-7TM ORF formed a clade with exclusively invertebrate sequences. Based on topology modelling with ee3, Sm-7TM was predicted to possess an intracellular C-terminal tail, which was expressed as a soluble thioredoxin fusion protein (Sm-7TMC) in Escherichia coli and purified using metal ion-affinity chromatography. A polyclonal antiserum against this domain was used to detect Sm-7TM in detergent-soluble parasite extracts and to immunolocalize the receptor to the tegument of adult S. mansoni.
Collapse
Affiliation(s)
- M. S. PEARSON
- Division of Infectious Diseases and Immunology, Queensland Institute of Medical Research and Australian Centre for International and Tropical Health and Nutrition, The University of Queensland, Brisbane, Queensland, Australia
| | - D. P. McMANUS
- Division of Infectious Diseases and Immunology, Queensland Institute of Medical Research and Australian Centre for International and Tropical Health and Nutrition, The University of Queensland, Brisbane, Queensland, Australia
| | - D. J. SMYTH
- Division of Infectious Diseases and Immunology, Queensland Institute of Medical Research and Australian Centre for International and Tropical Health and Nutrition, The University of Queensland, Brisbane, Queensland, Australia
| | - M. K. JONES
- Division of Infectious Diseases and Immunology, Queensland Institute of Medical Research and Australian Centre for International and Tropical Health and Nutrition, The University of Queensland, Brisbane, Queensland, Australia
| | - A. M. SYKES
- Division of Infectious Diseases and Immunology, Queensland Institute of Medical Research and Australian Centre for International and Tropical Health and Nutrition, The University of Queensland, Brisbane, Queensland, Australia
| | - A. LOUKAS
- Division of Infectious Diseases and Immunology, Queensland Institute of Medical Research and Australian Centre for International and Tropical Health and Nutrition, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
38
|
Nascimento EJM, Amorim RV, Cavalcanti A, Alves VF, Nakazawa M, Pereira VRA, Lucena-Silva N. Assessment of a DNA vaccine encoding an anchored-glycosylphosphatidylinositol tegumental antigen complexed to protamine sulphate on immunoprotection against murine schistosomiasis. Mem Inst Oswaldo Cruz 2007; 102:21-7. [PMID: 17293994 DOI: 10.1590/s0074-02762007000100003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2006] [Accepted: 11/29/2006] [Indexed: 11/22/2022] Open
Abstract
Protamine sulphate/DNA complexes have been shown to protect DNA from DNase digestion in a lipid system for gene transfer. A DNA-based vaccine complexed to protamine sulphate was used to induce an immune response against Schistosoma mansoni anchored-glycosylphosphatidylinositol tegumental antigen in BALB/c mice. The protection elicited ranged from 33 to 44%. The spectrum of the elicited immune response induced by the vaccine formulation without protamine was characterized by a high level of IgG (IgG1> IgG2a). Protamine sulphate added to the DNA vaccine formulation retained the green fluorescent protein encoding-plasmid longer in muscle and spleen. The experiments in vivo showed that under protamine sulphate effect, the scope of protection remained unchanged, but a modulation in antibody production (IgG1= IgG2a) was observed.
Collapse
|
39
|
Gene discovery for the carcinogenic human liver fluke, Opisthorchis viverrini. BMC Genomics 2007; 8:189. [PMID: 17587442 PMCID: PMC1913519 DOI: 10.1186/1471-2164-8-189] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2007] [Accepted: 06/22/2007] [Indexed: 12/30/2022] Open
Abstract
Background Cholangiocarcinoma (CCA) – cancer of the bile ducts – is associated with chronic infection with the liver fluke, Opisthorchis viverrini. Despite being the only eukaryote that is designated as a 'class I carcinogen' by the International Agency for Research on Cancer, little is known about its genome. Results Approximately 5,000 randomly selected cDNAs from the adult stage of O. viverrini were characterized and accounted for 1,932 contigs, representing ~14% of the entire transcriptome, and, presently, the largest sequence dataset for any species of liver fluke. Twenty percent of contigs were assigned GO classifications. Abundantly represented protein families included those involved in physiological functions that are essential to parasitism, such as anaerobic respiration, reproduction, detoxification, surface maintenance and feeding. GO assignments were well conserved in relation to other parasitic flukes, however, some categories were over-represented in O. viverrini, such as structural and motor proteins. An assessment of evolutionary relationships showed that O. viverrini was more similar to other parasitic (Clonorchis sinensis and Schistosoma japonicum) than to free-living (Schmidtea mediterranea) flatworms, and 105 sequences had close homologues in both parasitic species but not in S. mediterranea. A total of 164 O. viverrini contigs contained ORFs with signal sequences, many of which were platyhelminth-specific. Examples of convergent evolution between host and parasite secreted/membrane proteins were identified as were homologues of vaccine antigens from other helminths. Finally, ORFs representing secreted proteins with known roles in tumorigenesis were identified, and these might play roles in the pathogenesis of O. viverrini-induced CCA. Conclusion This gene discovery effort for O. viverrini should expedite molecular studies of cholangiocarcinogenesis and accelerate research focused on developing new interventions, drugs and vaccines, to control O. viverrini and related flukes.
Collapse
|
40
|
Loukas A, Tran M, Pearson MS. Schistosome membrane proteins as vaccines. Int J Parasitol 2006; 37:257-63. [PMID: 17222846 DOI: 10.1016/j.ijpara.2006.12.001] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2006] [Revised: 11/28/2006] [Accepted: 12/03/2006] [Indexed: 01/08/2023]
Abstract
Schistosomes are parasitic blood flukes that infect approximately 200 million people and are arguably the most important human helminth in terms of mortality. The outermost surface of intra-mammalian stages of the parasite, the tegument, is the key to the parasite's success, but it is also generally viewed as the most susceptible target for vaccines and drugs. Over the past 2 years the proteome of the Schistosoma mansoni tegument has been investigated and these studies revealed surprisingly few proteins that are predicted to be accessible to the host immune response, namely proteins with at least one membrane-spanning domain. However, of this handful of proteins, some are showing great promise as recombinant vaccines against schistosomiasis at a pre-clinical level. In particular, the tetraspanin family of integral membrane proteins appears to be abundantly represented in the tegument, and convergent data using the mouse vaccine model and correlates of protective immunity in naturally exposed people suggests that this family of membrane proteins offer great promise for schistosomiasis vaccines. With the recent advances in schistosome genomics and proteomics, a new suite of potential vaccine antigens are presented and these warrant detailed investigation and appropriate funding over the next few years.
Collapse
Affiliation(s)
- Alex Loukas
- Helminth Biology Laboratory, Division of Infectious Diseases and Immunology, Queensland Institute of Medical Research Brisbane, Qld 4006, Australia.
| | | | | |
Collapse
|
41
|
Abstract
The syncytial cytoplasmic layer, termed the tegument, which covers the entire surface of adult schistosomes, is a major interface between the parasite and its host. Since schistosomes can survive for decades within the host bloodstream, they are clearly able to evade host immune responses, and their ability is dependent on the properties of the tegument surface. We review here the molecular organization and biochemical functions of the tegument, combining the extensive literature over the last three decades with recent proteomic studies. We have interpreted the organization of the tegument surface as bounded by a conventional plasma membrane overlain by a membrane-like secretion, the membranocalyx, with which host molecules can associate. The range of parasite proteins, glycans and lipids found in the surface complex is evaluated, together with the host molecules detected. We consider the way in which the tegument surface is formed after cercarial penetration into the skin, and changes that occur as parasites develop to maturity. Lastly, we review the evidence on surface dynamics and turnover.
Collapse
Affiliation(s)
- Patrick J Skelly
- Tufts Cummings School of Veterinary Medicine, Department of Biomedical Sciences, 20 Westboro Road, North Grafton, MA 01536, USA
| | | |
Collapse
|
42
|
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: 1.2] [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
|
43
|
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
|
44
|
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: 1.4] [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
|
45
|
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: 3.4] [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
|
46
|
Cook RM, Carvalho-Queiroz C, Wilding G, LoVerde PT. Nucleic acid vaccination with Schistosoma mansoni antioxidant enzyme cytosolic superoxide dismutase and the structural protein filamin confers protection against the adult worm stage. Infect Immun 2004; 72:6112-24. [PMID: 15385516 PMCID: PMC517585 DOI: 10.1128/iai.72.10.6112-6124.2004] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Schistosomiasis remains a worldwide endemic cause of chronic and debilitating illness. There are two paradigms that exist in schistosome immunology. The first is that the schistosomule stages are the most susceptible to immune killing, and the second is that the adult stage, through evolution of defense mechanisms, can survive in the hostile host environment. One mechanism that seems to aid the adult worm in evading immune killing is the expression of antioxidant enzymes to neutralize the effects of reactive oxygen and nitrogen species. Here, we challenge one paradigm by targeting adult Schistosoma mansoni worms for immune elimination in an experimental mouse model using two S. mansoni antioxidants, cytosolic superoxide dismutase (SmCT-SOD) and glutathione peroxidase (SmGPX), and a partial coding sequence for a structural protein, filamin, as DNA vaccine candidates. DNA vaccination with SmCT-SOD induced a mean of 39% protection, filamin induced a mean of 50% protection, and SmGPX induced no protection compared to controls following challenge with adult worms by surgical transfer. B- and T-cell responses were analyzed in an attempt to define the protective immune mechanism(s) involved in adult worm killing. SmCT-SOD-immunized mice presented with a T1 response, and filamin-immunized mice showed a mixed T1-T2 response. We provide evidence for natural boosting after vaccination. Our results demonstrate that adult worms can be targeted for immune elimination through vaccination. This represents an advance in schistosome vaccinology and allows for the development of a therapeutic as well as a prophylactic vaccine.
Collapse
Affiliation(s)
- Rosemary M Cook
- Department of Microbiology and Immunology, School of Medicine and Biomedical Sciences, State University of New York, Buffalo, New York 14214, USA
| | | | | | | |
Collapse
|
47
|
Abstract
While there are many challenges in vaccine development, none is greater than that of developing vaccines against large metazoan parasites such as schistosomes, the parasitic worms that are responsible for schistosomiasis. Initial optimism stemming from the identification of the first vaccine candidate antigens that gave protection in animals has been dashed by the failure, as yet, of any of the vaccine candidate antigens to enter Phase III clinical trials. Now, despite an improved understanding of the biology of the parasites and of the immune responses they stimulate in naturally exposed populations, the vaccine effort is stalled. The control effort has switched heavily in favour of the wider use of conventional chemotherapy with praziquantel, which is now affordable by all but the poorest countries. Disagreements among researchers in the schistosome field as to whether or not a vaccine is needed have not helped convince funding agencies that schistosomiasis vaccines, rather than drugs, should be a priority. With the schistosome genome projects at an advanced stage plus the power of the proteomics, perhaps it is still too early to call time on schistosome vaccine development.
Collapse
Affiliation(s)
- Paul Hagan
- Division of Infection and Immunity, Joseph Black Building (B4-09d), Institute of Biomedical and Life Sciences, University of Glasgow, G12 8QQ UK.
| | | |
Collapse
|
48
|
Da'Dara AA, Skelly PJ, Walker CM, Harn DA. A DNA-prime/protein-boost vaccination regimen enhances Th2 immune responses but not protection following Schistosoma mansoni infection. Parasite Immunol 2004; 25:429-37. [PMID: 14651590 DOI: 10.1111/j.1365-3024.2003.00651.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
DNA immunization represents a promising vaccine strategy that has been reasonably successful, and will likely play an even greater role in vaccine development as these vaccines continue to be improved. We have developed a partially protective DNA vaccine against schistosome infection based on a 23-kDa integral membrane protein, Sm23. The focus of this study was to compare immunogenicity and efficacy of vaccination regimens utilizing Sm23 DNA vaccine alone vs. regimens that utilized both Sm23 DNA and Sm23 in recombinant protein form. We found that priming and boosting with the Sm23 DNA construct (Sm23-pcDNA) resulted in a significant level of protection against challenge infection (36-44%). In contrast, altering this protocol by changing the boost from Sm23 DNA to boosting with recombinant Sm23 protein (rSm23) formulated in aluminium hydroxide (alum) failed to induce a significant reduction in worm burdens. Similarly, mice primed and boosted with the rSm23 in alum also did not develop significant levels of protection against challenge infection. We hypothesize that the differences in the ability to drive protective immunity using the DNA prime-DNA boost strategy and the inability to do so when recombinant Sm23 in alum was substituted for Sm23 DNA is due to driving of different immune responses. In support of this, we found that mice primed and boosted with Sm23-pcDNA had Th1-type immune responses characterized by low anti-Sm23 IgG1 : IgG2a antibody isotype ratios, whereas mice boosted with rSm23 had higher IgG1 : IgG2a ratios. In addition, priming and boosting with rSm23 elicited mainly IgG1 antibodies with no detectable IgG2a, indicative of a polarized Th2-type immune response. Thus, similar to our earlier work, the results of this study show that protective vaccination using Sm23 is associated with a Th1 immune response, and efficacy is diminished using protocols that diminish this Th1 bias. In our study, this was likely due to the use of the Th2-driving adjuvant alum, and future studies are planned where we will compare the protective efficacy of rSm23 administered with Th1-type adjuvants.
Collapse
Affiliation(s)
- Akram A Da'Dara
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA 02115, USA.
| | | | | | | |
Collapse
|
49
|
Knox DP. Technological advances and genomics in metazoan parasites. Int J Parasitol 2004; 34:139-52. [PMID: 15037101 DOI: 10.1016/j.ijpara.2003.10.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2003] [Revised: 10/24/2003] [Accepted: 10/31/2003] [Indexed: 10/26/2022]
Abstract
Molecular biology has provided the means to identify parasite proteins, to define their function, patterns of expression and the means to produce them in quantity for subsequent functional analyses. Whole genome and expressed sequence tag programmes, and the parallel development of powerful bioinformatics tools, allow the execution of genome-wide between stage or species comparisons and meaningful gene-expression profiling. The latter can be undertaken with several new technologies such as DNA microarray and serial analysis of gene expression. Proteome analysis has come to the fore in recent years providing a crucial link between the gene and its protein product. RNA interference and ballistic gene transfer are exciting developments which can provide the means to precisely define the function of individual genes and, of importance in devising novel parasite control strategies, the effect that gene knockdown will have on parasite survival.
Collapse
Affiliation(s)
- D P Knox
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, Midlothian, Scotland EH26 0PZ, UK.
| |
Collapse
|
50
|
Shalaby KA, Yin L, Thakur A, Christen L, Niles EG, LoVerde PT. Protection against Schistosoma mansoni utilizing DNA vaccination with genes encoding Cu/Zn cytosolic superoxide dismutase, signal peptide-containing superoxide dismutase and glutathione peroxidase enzymes. Vaccine 2003; 22:130-6. [PMID: 14604580 DOI: 10.1016/s0264-410x(03)00535-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Protection against Schistosoma mansoni infection in C57BL/6 female mice was evaluated by two DNA vaccination strategies. Mice were either vaccinated by intramuscular injection with pcDNAI/Amp constructs encoding either Cu/Zn cytosolic superoxide dismutase (CT-SOD), signal peptide-containing SOD (SP-SOD), glutathione peroxidase (GPX(bb)) or a mutated form of GPX (GPX(m)), or primed with naked DNA constructs and boosted with recombinant vaccinia virus (RVV) containing the same genes. Animals were then challenged with S. mansoni and the level of protection was assessed as the reduction in worm burden. CT-SOD showed significant levels of protection compared to the control group, ranging between 44 and 60%, while SP-SOD exhibited from 22 to 45%. GPX(bb) showed levels of protection (23-55%) higher than GPX(m) (25-34%). The prime-boost strategy gave the same results as naked DNA or recombinant vaccinia virus alone except in the case of GPX, where the protection was 85%.
Collapse
Affiliation(s)
- Kamal A Shalaby
- Department of Microbiology, and Immunology, School of Medicine and Biomedical Sciences, State University of New York, 138 Farber Hall, Buffalo, NY 14214, USA
| | | | | | | | | | | |
Collapse
|