1
|
Minutti AF, Sasse JP, Dos Santos Silva AC, Martins TA, Martinez V, de Souza Lima Nino B, de Souza Rodrigues F, de Barros LD, Garcia JL. Evaluation of a DNA vector plasmid encoding a partial rop18 gene from toxoplasma gondii in domestic cats as a vaccine candidate. Vaccine 2025; 54:126965. [PMID: 40054140 DOI: 10.1016/j.vaccine.2025.126965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/14/2025] [Accepted: 02/25/2025] [Indexed: 05/13/2025]
Abstract
The present study aimed to evaluate a DNA vector plasmid encoding a partial rop18 gene from Toxoplasma gondii in domestic cats as a potential vaccine candidate. Four domestic cats (Felis catus) were used, of which two animals received 25 μg of pcDNA 3.1 + rop18, and two received 25 μg of pcDNA 3.1. All animals received intramuscular immunizations with four doses every three weeks along with 1.5 % levamisole. Thirty days after the last immunization, the animals were infected with 300 tissue cysts from ToxoDB #182 strain, a non-archetypal genotype isolated from a wild cat. Fecal examinations were performed for oocyst shedding. Enzyme-linked immunosorbent assay and western blotting analyses with recombinant ROP18 were performed to assess the humoral immune response. Animals that received plasmid containing the partial T. gondii rop18 gene produced specific IgG antibodies and shed 53.3 % fewer oocysts than controls. The two groups of animals showed no statistically significant differences (p > 0.05) in oocyst shedding; however, they showed significant differences in the detection of anti-Toxoplasma antibodies (p < 0.05). In conclusion, the T. gondii rop18 gene is a potential vaccine candidate against oocyst shedding in cats.
Collapse
Affiliation(s)
- Ana Flávia Minutti
- Department of Preventive Veterinary Medicine, Laboratory of Animal Protozoology, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, PR 445 km 380, 86057-970 Londrina, PR, Brazil
| | - João Pedro Sasse
- Department of Preventive Veterinary Medicine, Laboratory of Animal Protozoology, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, PR 445 km 380, 86057-970 Londrina, PR, Brazil
| | - Ana Clécia Dos Santos Silva
- Department of Preventive Veterinary Medicine, Laboratory of Animal Protozoology, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, PR 445 km 380, 86057-970 Londrina, PR, Brazil
| | - Thais Agostinho Martins
- Department of Preventive Veterinary Medicine, Laboratory of Animal Protozoology, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, PR 445 km 380, 86057-970 Londrina, PR, Brazil
| | - Valentina Martinez
- Department of Preventive Veterinary Medicine, Laboratory of Animal Protozoology, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, PR 445 km 380, 86057-970 Londrina, PR, Brazil
| | - Beatriz de Souza Lima Nino
- Department of Preventive Veterinary Medicine, Laboratory of Animal Protozoology, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, PR 445 km 380, 86057-970 Londrina, PR, Brazil
| | - Fernando de Souza Rodrigues
- Department of Preventive Veterinary Medicine, Laboratory of Animal Protozoology, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, PR 445 km 380, 86057-970 Londrina, PR, Brazil
| | - Luiz Daniel de Barros
- Laboratory of Veterinary Parasitology and Parasitic Diseases, Universidade Federal de Lavras - UFLA, Lavras, MG, Brazil
| | - João Luis Garcia
- Department of Preventive Veterinary Medicine, Laboratory of Animal Protozoology, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid, PR 445 km 380, 86057-970 Londrina, PR, Brazil.
| |
Collapse
|
2
|
Daher D, Shaghlil A, Sobh E, Hamie M, Hassan ME, Moumneh MB, Itani S, El Hajj R, Tawk L, El Sabban M, El Hajj H. Comprehensive Overview of Toxoplasma gondii-Induced and Associated Diseases. Pathogens 2021; 10:pathogens10111351. [PMID: 34832507 PMCID: PMC8625914 DOI: 10.3390/pathogens10111351] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/07/2021] [Accepted: 10/14/2021] [Indexed: 12/03/2022] Open
Abstract
Toxoplasma gondii (T. gondii) is a prevalent protozoan parasite of medical and veterinary significance. It is the etiologic agent of toxoplasmosis, a neglected disease in which incidence and symptoms differ between patients and regions. In immunocompetent patients, toxoplasmosis manifests as acute and chronic forms. Acute toxoplasmosis presents as mild or asymptomatic disease that evolves, under the host immune response, into a persistent chronic disease in healthy individuals. Chronic toxoplasmosis establishes as latent tissue cysts in the brain and skeletal muscles. In immunocompromised patients, chronic toxoplasmosis may reactivate, leading to a potentially life-threatening condition. Recently, the association between toxoplasmosis and various diseases has been shown. These span primary neuropathies, behavioral and psychiatric disorders, and different types of cancer. Currently, a direct pre-clinical or clinical molecular connotation between toxoplasmosis and most of its associated diseases remains poorly understood. In this review, we provide a comprehensive overview on Toxoplasma-induced and associated diseases with a focus on available knowledge of the molecular players dictating these associations. We will also abridge the existing therapeutic options of toxoplasmosis and highlight the current gaps to explore the implications of toxoplasmosis on its associated diseases to advance treatment modalities.
Collapse
Affiliation(s)
- Darine Daher
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (D.D.); (M.H.); (M.E.H.); (M.B.M.); (S.I.)
| | - Ahmad Shaghlil
- Department of Biology, Faculty of Sciences, R. Hariri Campus, Lebanese University, Beirut 1107 2020, Lebanon; (A.S.); (E.S.)
| | - Eyad Sobh
- Department of Biology, Faculty of Sciences, R. Hariri Campus, Lebanese University, Beirut 1107 2020, Lebanon; (A.S.); (E.S.)
| | - Maguy Hamie
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (D.D.); (M.H.); (M.E.H.); (M.B.M.); (S.I.)
| | - Malika Elhage Hassan
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (D.D.); (M.H.); (M.E.H.); (M.B.M.); (S.I.)
| | - Mohamad Bahij Moumneh
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (D.D.); (M.H.); (M.E.H.); (M.B.M.); (S.I.)
| | - Shaymaa Itani
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (D.D.); (M.H.); (M.E.H.); (M.B.M.); (S.I.)
| | - Rana El Hajj
- Department of Biological Sciences, Beirut Arab University, Beirut 1107 2809, Lebanon;
| | - Lina Tawk
- Department of Medical Laboratory Sciences, Faculty of Health Sciences, University of Balamand, Beirut 1100 2807, Lebanon;
| | - Marwan El Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon;
| | - Hiba El Hajj
- Department of Experimental Pathology, Microbiology and Immunology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon; (D.D.); (M.H.); (M.E.H.); (M.B.M.); (S.I.)
- Correspondence: ; Tel.: +961–1-350000 (ext. 4897)
| |
Collapse
|
3
|
Beck B, Grochow T, Schares G, Blaga R, Le Roux D, Bangoura B, Daugschies A, Fietz SA. Burden and regional distribution of Toxoplasma gondii cysts in the brain of COBB 500 broiler chickens following chronic infection with 76K strain. Vet Parasitol 2021; 296:109497. [PMID: 34147768 DOI: 10.1016/j.vetpar.2021.109497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 10/21/2022]
Abstract
Toxoplasmosis is a worldwide zoonosis caused by the obligate intracellular apicomplexan parasite Toxoplasma gondii (T. gondii). Chickens are ground-feeders and represent, especially if free-range, important intermediate hosts in the epidemiology of toxoplasmosis and are used as sentinels of environmental contamination with T. gondii oocysts. Until now, little is known about the burden and regional distribution of T. gondii cysts in the chicken brain. It was therefore the aim of this study to investigate the abundance and specific distribution of T. gondii cysts within the chicken brain following chronic infection with a type II strain (76 K) of T. gondii. A total of 29 chickens were included in the study and divided into control group (n = 9) and two different infection groups, a low dose (n = 10) and a high dose (n = 10) group, which were orally inoculated with 1500 or 150,000 T. gondii oocysts per animal, respectively. Seroconversion was detected in the majority of chickens of the high dose group, but not in the animals of the low dose and the control group. Moreover, T. gondii DNA was detected most frequently in the brain and more frequently in the heart than in liver, spleen, thigh and pectoral muscle using qPCR analysis. The number of T. gondii cysts, quantified in the chicken brain using histological analysis, seems to be considerably lower as compared to studies in rodents, which might explain why T. gondii infected chickens very rarely, if at all, develop neurological deficits. Similar to observations in mice, in which no lateralisation for T. gondii cysts was reported, T. gondii cysts were distributed nearly equally between the left and right chicken brain hemispheres. When different brain regions (fore-, mid- and hindbrain) were compared, all T. gondii cysts were located in the forebrain with the overwhelming majority of these cysts being present in the telencephalic pallium and subpallium. More studies including different strains and higher doses of T. gondii are needed in order to precisely evaluate its cyst burden and regional distribution in the chicken brain. Together, our findings provide insights into the course of T. gondii infection in chickens and are important to understand the differences of chronic T. gondii infection in the chicken and mammalian brain.
Collapse
Affiliation(s)
- Britta Beck
- Institute of Parasitology, Centre for Infectious Diseases, Faculty of Veterinary Medicine, University of Leipzig, 04103, Leipzig, Germany; Institute of Veterinary Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Leipzig, 04103, Leipzig, Germany
| | - Thomas Grochow
- Institute of Parasitology, Centre for Infectious Diseases, Faculty of Veterinary Medicine, University of Leipzig, 04103, Leipzig, Germany; Institute of Veterinary Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Leipzig, 04103, Leipzig, Germany
| | - Gereon Schares
- Friedrich-Loeffler-Institut, Institute of Epidemiology, 17493, Greifswald-Insel Riems, Germany
| | - Radu Blaga
- Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, F-94700, France
| | - Delphine Le Roux
- Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, F-94700, France
| | - Berit Bangoura
- Wyoming State Veterinary Laboratory, Department of Veterinary Sciences, University of Wyoming, Laramie, WY, 82070, USA
| | - Arwid Daugschies
- Institute of Parasitology, Centre for Infectious Diseases, Faculty of Veterinary Medicine, University of Leipzig, 04103, Leipzig, Germany
| | - Simone A Fietz
- Institute of Veterinary Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Leipzig, 04103, Leipzig, Germany.
| |
Collapse
|
4
|
Pagheh AS, Sarvi S, Sharif M, Rezaei F, Ahmadpour E, Dodangeh S, Omidian Z, Hassannia H, Mehrzadi S, Daryani A. Toxoplasma gondii surface antigen 1 (SAG1) as a potential candidate to develop vaccine against toxoplasmosis: A systematic review. Comp Immunol Microbiol Infect Dis 2020; 69:101414. [PMID: 31958746 DOI: 10.1016/j.cimid.2020.101414] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 12/30/2019] [Accepted: 12/31/2019] [Indexed: 12/13/2022]
Abstract
Toxoplasma gondii is an intracellular parasite that infects a broad range of animal species and humans. As the main surface antigen of the tachyzoite, SAG1 is involved in the process of recognition, adhesion and invasion of host cells. The aim of the current systematic review study is to clarify the latest status of studies in the literature regarding SAG1-associated recombinant proteins or SAG1-associated recombinant DNAs as potential vaccines against toxoplasmosis. Data were systematically collected from six databases including PubMed, Science Direct, Web of Science, Google Scholar, EBSCO and Scopus, up to 1st of January 2019. A total of 87 articles were eligible for inclusion criteria in the current systematic review. The most common antigens used for experimental cocktail vaccines together with SAG1 were ROP2 and SAG2. In addition, the most parasite strains used were RH and ME49. Freund's adjuvant and cholera toxin have been predominantly utilized. Furthermore, regarding the animal models, route and dose of vaccination, challenge methods, measurement of immune responses and cyst burden have been discussed in the text. Most of these experimental vaccines induce immune responses and have a high degree of protection against parasite infections, increase survival rates and duration and reduce cyst burdens. The data demonstrated that SAG1 antigen has a high potential for use as a vaccine and provided a promising approach for protecting humans and animals against toxoplasmosis.
Collapse
Affiliation(s)
- Abdol Sattar Pagheh
- Infectious Disease Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Shahabeddin Sarvi
- Toxoplasmosis Research Center, Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mehdi Sharif
- Department of Parasitology, School of Medicine, Sari Branch, Islamic AZAD University, Sari, Iran
| | - Fatemeh Rezaei
- Toxoplasmosis Research Center, Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ehsan Ahmadpour
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Parasitology and Mycology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samira Dodangeh
- Toxoplasmosis Research Center, Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zahra Omidian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Hadi Hassannia
- Immunonogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ahmad Daryani
- Toxoplasmosis Research Center, Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran..
| |
Collapse
|
5
|
Duan Q, Xia P, Nandre R, Zhang W, Zhu G. Review of Newly Identified Functions Associated With the Heat-Labile Toxin of Enterotoxigenic Escherichia coli. Front Cell Infect Microbiol 2019; 9:292. [PMID: 31456954 PMCID: PMC6700299 DOI: 10.3389/fcimb.2019.00292] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 07/29/2019] [Indexed: 12/11/2022] Open
Abstract
Heat-labile toxin (LT) is a well-characterized powerful enterotoxin produced by enterotoxigenic Escherichia coli (ETEC). This toxin is known to contribute to diarrhea in young children in developing countries, international travelers, as well as many different species of young animals. Interestingly, it has also been revealed that LT is involved in other activities in addition to its role in enterotoxicity. Recent studies have indicated that LT toxin enhances enteric pathogen adherence and subsequent intestinal colonization. LT has also been shown to act as a powerful adjuvant capable of upregulating vaccine antigenicity; it also serves as a protein or antigenic peptide display platform for new vaccine development, and can be used as a naturally derived cell targeting and protein delivery tool. This review summarizes the epidemiology, secretion, delivery, and mechanisms of action of LT, while also highlighting new functions revealed by recent studies.
Collapse
Affiliation(s)
- Qiangde Duan
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Pengpeng Xia
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Rahul Nandre
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
| | - Weiping Zhang
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Guoqiang Zhu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
6
|
Watson GF, Davis PH. Systematic review and meta-analysis of variation in Toxoplasma gondii cyst burden in the murine model. Exp Parasitol 2019; 196:55-62. [PMID: 30562481 PMCID: PMC6447088 DOI: 10.1016/j.exppara.2018.12.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/14/2018] [Accepted: 12/14/2018] [Indexed: 11/20/2022]
Abstract
Toxoplasma gondii is an obligate intracellular protozoan parasite that infects approximately 30% of the population of the United States, with worldwide distribution. The chronic (latent) infection, mediated by the bradyzoite parasite life stage, has attracted attention due to possible links to host behavioral alteration and psychomotor effects. Mice are a common model organism for studying the chronic stage, as they are natural hosts of infection. Notably, published studies demonstrate vast ranges of measured cyst burden within the murine brain tissue. The inconsistency of measured cyst burden within and between experiments makes interpretation of statistical significance difficult, potentially confounding studies of experimental anti-parasitic approaches. This review analyzes variation in measured cyst burden in a wide array of experimental mouse infections across published literature. Factors such as parasite infection strain, mouse strain, mode of infection, and infectious dose were all examined. The lowest variation in measured cyst burden occurred with the commonly available Balb/c and CBA mice undergoing infection by the ME49 strain of T. gondii. A summary of cyst variation and average cyst counts in T. gondii mouse models is presented, which may be useful for designing future experiments.
Collapse
Affiliation(s)
| | - Paul H Davis
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, USA.
| |
Collapse
|
7
|
A systematic review of Toxoplasma gondii antigens to find the best vaccine candidates for immunization. Microb Pathog 2018; 126:172-184. [PMID: 30399440 DOI: 10.1016/j.micpath.2018.11.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/30/2018] [Accepted: 11/02/2018] [Indexed: 11/24/2022]
Abstract
At present, there is not any available accepted vaccine for prevention of Toxoplasma gondii (T. gondii) in human and animals. We conducted literature search through English (Google Scholar, PubMed, Science Direct, Scopus, EBSCO, ISI Web of Science) scientific paper databases to find the best vaccine candidates against toxoplasmosis among T. gondii antigens. Articles with information on infective stage, pathogenicity, immunogenicity and characterization of antigens were selected. We considered that the ideal and significant vaccines should include different antigens and been expressed in all infective stages of the parasite with a high pathogenicity and immunogenicity. Evaluation within this systematic review indicates that MIC 3, 4, 13, ROP 2, RON 5, GRA 1, 6, 8, 14 are expressed in all three infective stages and have pathogenicity and immunogenicity. MIC 5, ROM 4, GRA 2, 4, 15, ROP 5, 16, 17, 38, RON 4, MIC 1, GRA 10, 12, 16, SAG 3 are expressed in only tachyzoites and bradyzoites stages of T. gondii with pathogenicity/immunogenicity. Some antigens appeared to be expressed in a single stage (tachyzoites) but have high pathogenicity and induce immune response. They include enolase2 (ENO2), SAG 1, SAG5D, HSP 70, ROM 1, ROM 5, AMA 1, ROP 18, RON2 and GRA 24. In conclusion, current vaccination against T. gondii infection is not satisfactory, and with the increasing number of high-risk individuals, the development of an effective and safe specific vaccine is greatly valuable for toxoplasmosis prevention. This systematic review reveals prepare candidates for immunization studies.
Collapse
|
8
|
ADP-ribosylating enterotoxins as vaccine adjuvants. Curr Opin Pharmacol 2018; 41:42-51. [PMID: 29702466 DOI: 10.1016/j.coph.2018.03.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 03/30/2018] [Indexed: 01/18/2023]
Abstract
Most infections are caused by pathogens that access the body at mucosal sites. Hence, development of mucosal vaccines to prevent local infection or invasion of pathogens appears highly warranted, especially since only mucosal immunization will stimulate strong local IgA responses and tissue resident memory CD4 and CD8 T cells. The most significant obstacle to developing such vaccines is the lack of approved adjuvants that can effectively and safely enhance relevant mucosal and systemic immune responses. The most potent mucosal adjuvants known today are the adenosine diphosphate (ADP)-ribosylating bacterial enterotoxins cholera toxin (CT) and Escherichia coli heat-labile toxins (LTs). Unfortunately, these molecules are also very toxic, which precludes their clinical use. However, much effort has been devoted to developing derivatives of these enterotoxins with low or no toxicity and retained adjuvant activity. Although it is fair to say that we know more about how these toxins affect the immune system than ever before, we still lack a detailed understanding of how and why these toxins are effective adjuvants. In the present review, we provide a state-of-the-art overview of the mechanism of action of the holotoxins and the strategies used for improving the toxin-based adjuvants.
Collapse
|
9
|
Lakhrif Z, Moreau A, Hérault B, Di-Tommaso A, Juste M, Moiré N, Dimier-Poisson I, Mévélec MN, Aubrey N. Targeted Delivery of Toxoplasma gondii Antigens to Dendritic Cells Promote Immunogenicity and Protective Efficiency against Toxoplasmosis. Front Immunol 2018. [PMID: 29515595 PMCID: PMC5826183 DOI: 10.3389/fimmu.2018.00317] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Toxoplasmosis is a major public health problem and the development of a human vaccine is of high priority. Efficient vaccination against Toxoplasma gondii requires both a mucosal and systemic Th1 immune response. Moreover, dendritic cells play a critical role in orchestrating the innate immune functions and driving specific adaptive immunity to T. gondii. In this study, we explore an original vaccination strategy that combines administration via mucosal and systemic routes of fusion proteins able to target the major T. gondii surface antigen SAG1 to DCs using an antibody fragment single-chain fragment variable (scFv) directed against DEC205 endocytic receptor. Our results show that SAG1 targeting to DCs by scFv via intranasal and subcutaneous administration improved protection against chronic T. gondii infection. A marked reduction in brain parasite burden is observed when compared with the intranasal or the subcutaneous route alone. DC targeting improved both local and systemic humoral and cellular immune responses and potentiated more specifically the Th1 response profile by more efficient production of IFN-γ, interleukin-2, IgG2a, and nasal IgA. This study provides evidence of the potential of DC targeting for the development of new vaccines against a range of Apicomplexa parasites.
Collapse
|
10
|
Yang WB, Zhou DH, Zou Y, Chen K, Liu Q, Wang JL, Zhu XQ, Zhao GH. Vaccination with a DNA vaccine encoding Toxoplasma gondii ROP54 induces protective immunity against toxoplasmosis in mice. Acta Trop 2017; 176:427-432. [PMID: 28935555 DOI: 10.1016/j.actatropica.2017.09.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/28/2017] [Accepted: 09/16/2017] [Indexed: 12/22/2022]
Abstract
Toxoplasma gondii is an obligatory intracellular protozoan, which infects most of the warm-blooded animals, causing serious public health problems and enormous economic losses worldwide. The rhoptry effector protein 54 (ROP54) has been indicated as a virulence factor that promotes Toxoplasma infection by modulating GBP2 loading onto parasite-containing vacuoles, which can modulate some aspects of the host immune response. In order to evaluate the immuno-protective value of ROP54, we constructed a eukaryotic recombinant plasmid expressing T. gondii ROP54 and intramuscularly immunized Kunming mice with this recombinant plasmid against acute and chronic toxoplasmosis. All mice immunized with pVAX-ROP54 elicited a high level of specific antibody responses, a significant increase of lymphocyte proliferation, and a significant level of Th1-type cytokines (IFN-γ, IL-2 and IL-12p70), in addition to an increased production of Th2-type cytokines (IL-4 and IL-10). These results demonstrated that pVAX-ROP54 induced significant cellular and humoral (Th1/Th2) immune responses, which extended the survival time (13.0±1.15days for pVAX-ROP54 vs 6.7±0.48days for pVAX I, 6.8±0.42days for PBS and 6.5±0.53 for blank control) and significantly reduced cyst burden (35.9% for pVAX-ROP54, 1% for pVAX I and 2% for PBS, compared with blank control) of immunized mice. These results indicate that the recombinant ROP54 plasmid can provide partial protection and might be a potential vaccine candidate against acute and chronic toxoplasmosis.
Collapse
|
11
|
Toxoplasma gondii : Immunological response of sheep to injections of recombinant SAG1, SAG2, GRA1 proteins coupled to the non-toxic microparticle muramyl dipeptide. Small Rumin Res 2017. [DOI: 10.1016/j.smallrumres.2017.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
12
|
Czarnewski P, Araújo ECB, Oliveira MC, Mineo TWP, Silva NM. Recombinant TgHSP70 Immunization Protects against Toxoplasma gondii Brain Cyst Formation by Enhancing Inducible Nitric Oxide Expression. Front Cell Infect Microbiol 2017; 7:142. [PMID: 28487847 PMCID: PMC5403831 DOI: 10.3389/fcimb.2017.00142] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/06/2017] [Indexed: 11/17/2022] Open
Abstract
Toxoplasma gondii is known to cause congenital infection in humans and animals and severe disease in immunocompromised individuals; consequently development of vaccines against the parasite is highly necessary. Under stress conditions, T. gondii expresses the highly immunogenic heat shock protein 70 (TgHSP70). Here, we assessed the protective efficacy of rTgHSP70 immunization combined with Alum in oral ME-49 T. gondii infection and the mechanisms involved on it. It was observed that immunized mice with rTgHSP70 or rTgHSP70 adsorbed in Alum presented a significantly reduced number of cysts in the brain that was associated with increased iNOS+ cell numbers in the organ, irrespective the use of the adjuvant. Indeed, ex vivo experiments showed that peritoneal macrophages pre-stimulated with rTgHSP70 presented increased NO production and enhanced parasite killing, and the protein was able to directly stimulate B cells toward antibody producing profile. In addition, rTgHSP70 immunization leads to high specific antibody titters systemically and a mixed IgG1/IgG2a response, with predominance of IgG1 production. Nonetheless, it was observed that the pretreatment of the parasite with rTgHSP70 immune sera was not able to control T. gondii internalization and replication by NIH fibroblast neither peritoneal murine macrophages, nor anti-rTgHSP70 antibodies were able to kill T. gondii by complement-mediated lysis, suggesting that these mechanisms are not crucial to resistance. Interestingly, when in combination with Alum, rTgHSP70 immunization was able to reduce inflammation in the brain of infected mice and in parallel anti-rTgHSP70 immune complexes in the serum. In conclusion, immunization with rTgHSP70 induces massive amounts of iNOS expression and reduced brain parasitism, suggesting that iNOS expression and consequently NO production in the brain is a protective mechanism induced by TgHSP70 immunization, therefore rTgHSP70 can be a good candidate for vaccine development against toxoplasmosis.
Collapse
Affiliation(s)
- Paulo Czarnewski
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of UberlândiaUberlândia, Brazil
| | - Ester C B Araújo
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of UberlândiaUberlândia, Brazil
| | - Mário C Oliveira
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of UberlândiaUberlândia, Brazil
| | - Tiago W P Mineo
- Laboratory of Immunoparasitology, Institute of Biomedical Sciences, Federal University of UberlândiaUberlândia, Brazil
| | - Neide M Silva
- Laboratory of Immunopathology, Institute of Biomedical Sciences, Federal University of UberlândiaUberlândia, Brazil
| |
Collapse
|
13
|
Zulpo DL, Igarashi M, Sammi AS, Santos JRD, Sasse JP, Cunha IALD, Taroda A, Barros LDD, Almeida JCD, Jenkins MC, Navarro IT, Garcia JL. rROP2 from Toxoplasma gondii as a potential vaccine against oocyst shedding in domestic cats. REVISTA BRASILEIRA DE PARASITOLOGIA VETERINARIA 2017; 26:67-73. [DOI: 10.1590/s1984-29612017007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/07/2017] [Indexed: 11/21/2022]
Abstract
Abstract The aim of the present study was to evaluate oocyst shedding in cats immunized by nasal route with T. gondii proteins ROP2. Twelve short hair cats (Felis catus) were divided in three groups G1, G2 and G3 (n=4). Animals from G1 received 100 μg of rROP2 proteins plus 20 μg of Quil-A, G2 received 100 μg of BSA plus 20 μg of Quil-A, and the G3 only saline solution (control group). All treatments were done by intranasal route at days 0, 21, 42, and 63. The challenge was performed in all groups on day 70 with ≅ 800 tissue cysts of ME-49 strain by oral route. Animals from G1 shed less oocysts (86.7%) than control groups. ELISA was used to detect anti-rROP2 IgG and IgA, however, there were no correlation between number of oocyst shedding by either IgG or IgA antibody levels. In the present work, in spite of lesser oocysts production in immunized group than control groups, it was not possible to associate the use of rROP2 via nostrils with protection against oocyst shedding. For the future, the use of either other recombinant proteins or DNA vaccine, in combination with rROP2 could be tested to try improving the efficacy of this kind of vaccine.
Collapse
Affiliation(s)
- Dauton Luiz Zulpo
- Universidade Estadual de Londrina, Brasil; Pontifícia Universidade Católica do Paraná, Brasil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Rashid I, Moiré N, Héraut B, Dimier-Poisson I, Mévélec MN. Enhancement of the protective efficacy of a ROP18 vaccine against chronic toxoplasmosis by nasal route. Med Microbiol Immunol 2016; 206:53-62. [PMID: 27757545 DOI: 10.1007/s00430-016-0483-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 10/11/2016] [Indexed: 10/20/2022]
Abstract
Infection with the parasite Toxoplasma gondii causes serious public health problems and is of great economic importance worldwide. No vaccine is currently available, so the design of efficient vaccine strategies is still a topical question. In this study, we evaluated the immunoprophylactic potential of a T. gondii virulence factor, the rhoptry kinase ROP18, in a mouse model of chronic toxoplasmosis: first using a recombinant protein produced in Schneider insect cells adjuvanted with poly I:C emulsified in Montanide SV71 by a parenteral route or adjuvanted with cholera toxin by the nasal route and second using a DNA plasmid encoding ROP18 adjuvanted with GM-CSF ± IL-12 DNA. If both intranasal and subcutaneous recombinant ROP18 immunizations induced predominantly anti-ROP18 IgG1 antibodies and generated a mixed systemic Th1-/Th2-type cellular immune response characterized by the production of IFN-γ, IL-2, Il-10 and IL-5, only intranasal vaccination induced a mucosal (IgA) humoral response in intestinal washes associated with a significant brain cyst reduction (50 %) after oral challenge with T. gondii cysts. DNA immunization induced antibodies and redirected the cellular immune response toward a Th1-type response (production of IFN-γ and IL-2) but did not confer protection. These results suggest that ROP18 could be a component of a subunit vaccine against toxoplasmosis and that strategies designed to enhance mucosal protective immune responses could lead to more encouraging results.
Collapse
Affiliation(s)
- Imran Rashid
- ISP, INRA, Université de Tours, UMR 1282, 37380, Nouzilly, France
| | - Nathalie Moiré
- ISP, INRA, Université de Tours, UMR 1282, 37380, Nouzilly, France
| | - Bruno Héraut
- ISP, INRA, Université de Tours, UMR 1282, 37380, Nouzilly, France
| | | | | |
Collapse
|
15
|
Lim SSY, Othman RY. Recent advances in Toxoplasma gondii immunotherapeutics. THE KOREAN JOURNAL OF PARASITOLOGY 2014; 52:581-93. [PMID: 25548409 PMCID: PMC4277020 DOI: 10.3347/kjp.2014.52.6.581] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 08/21/2014] [Accepted: 08/22/2014] [Indexed: 12/11/2022]
Abstract
Toxoplasmosis is an opportunistic infection caused by the protozoan parasite Toxoplasma gondii. T. gondii is widespread globally and causes severe diseases in individuals with impaired immune defences as well as congenitally infected infants. The high prevalence rate in some parts of the world such as South America and Africa, coupled with the current drug treatments that trigger hypersensitivity reactions, makes the development of immunotherapeutics intervention a highly important research priority. Immunotherapeutics strategies could either be a vaccine which would confer a pre-emptive immunity to infection, or passive immunization in cases of disease recrudescence or recurrent clinical diseases. As the severity of clinical manifestations is often greater in developing nations, the development of well-tolerated and safe immunotherapeutics becomes not only a scientific pursuit, but a humanitarian enterprise. In the last few years, much progress has been made in vaccine research with new antigens, novel adjuvants, and innovative vaccine delivery such as nanoparticles and antigen encapsulations. A literature search over the past 5 years showed that most experimental studies were focused on DNA vaccination at 52%, followed by protein vaccination which formed 36% of the studies, live attenuated vaccinations at 9%, and heterologous vaccination at 3%; while there were few on passive immunization. Recent progress in studies on vaccination, passive immunization, as well as insights gained from these immunotherapeutics is highlighted in this review.
Collapse
Affiliation(s)
- Sherene Swee-Yin Lim
- Genetics and Molecular Biology Department, Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Rofina Yasmin Othman
- Genetics and Molecular Biology Department, Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia. ; Centre for Research in Biotechnology for Agriculture, University of Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
16
|
Prandota J. Possible link between Toxoplasma gondii and the anosmia associated with neurodegenerative diseases. Am J Alzheimers Dis Other Demen 2014; 29:205-14. [PMID: 24413543 PMCID: PMC10852608 DOI: 10.1177/1533317513517049] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Toxoplasma gondii is an intracellular protozoan infecting 30% to 50% of global human population. Recently, it was suggested that chronic latent neuroinflammation caused by the parasite may be responsible for the development of several neurodegenerative diseases manifesting with the loss of smell. Studies in animals inoculated with the parasite revealed cysts in various regions of the brain, including olfactory bulb. Development of behavioral changes was paralleled by the preferential persistence of cysts in defined anatomic structures of the brain, depending on the host, strain of the parasite, its virulence, and route of inoculation. Olfactory dysfunction reported in Alzheimer's disease, multiple sclerosis, and schizophrenia was frequently associated with the significantly increased serum anti-T gondii immunoglobulin G antibody levels. Damage of the olfactory system may be also at least in part responsible for the development of depression because T gondii infection worsened mood in such patients, and the olfactory bulbectomized rat serves as a model of depression.
Collapse
Affiliation(s)
- Joseph Prandota
- Department of Social Pediatrics, Faculty of Health Sciences, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
17
|
Zheng B, Lu S, Tong Q, Kong Q, Lou D. The virulence-related rhoptry protein 5 (ROP5) of Toxoplasma Gondii is a novel vaccine candidate against toxoplasmosis in mice. Vaccine 2013; 31:4578-84. [PMID: 23928460 DOI: 10.1016/j.vaccine.2013.07.058] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 06/29/2013] [Accepted: 07/25/2013] [Indexed: 12/22/2022]
Abstract
Infections with the intracellular protozoan parasite Toxoplasma gondii pose a serious public health problem and are of great economic importance worldwide. The parasite rhoptry protein 5 (ROP5) has been implicated as a major virulence factor that reduces the accumulation of immunity-related GTPases (IRG) in parasitophorous vacuole membrane (PVM), which maintains PVM integrity and evades IFNγ-mediated killing by intracellular parasites. To study the immunoprotective value of ROP5, BALB/c mice were immunized with a recombinant form of the protein administered alone or in combination with another promising vaccine antigen, rSAG1. All mice vaccinated with the recombinant antigens developed a high level of specific antibody responses against soluble tachyzoite antigens (STAg), a statistically significant increase of the splenocyte proliferation response, and significant levels of IFN-γ and IL-2 production. In contrast to rSAG1, which only stimulated the release of IFN-γ and IL-2, rROP5 induced the specific production of IL-10, the Th2-type cytokine, in addition to IFN-γ and IL-2. These results demonstrated that rROP5 could induce significant cellular and humoral (Th1/Th2) immune responses. Moreover, mice immunized with rROP5 displayed a prolonged survival time against a lethal challenge with the T. gondii RH strain. Additionally, vaccination with the mixture of rROP5+rSAG1 resulted in higher levels of T. gondii-specific IgG antibodies and lymphocyte proliferative responses and conferred more efficient protection against T. gondii challenge compared to immunization with rROP5 or rSAG1 alone. Our studies show that recombinant ROP5 antigen may be a promising vaccine candidate against toxoplasmosis. To our knowledge, this is the first report to evaluate the immunoprotective value of ROP5.
Collapse
Affiliation(s)
- Bin Zheng
- Institute of Parasitic Diseases, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, PR China
| | | | | | | | | |
Collapse
|
18
|
Mendes ÉA, Fonseca FG, Casério BM, Colina JP, Gazzinelli RT, Caetano BC. Recombinant vaccines against T. gondii: comparison between homologous and heterologous vaccination protocols using two viral vectors expressing SAG1. PLoS One 2013; 8:e63201. [PMID: 23690999 PMCID: PMC3654925 DOI: 10.1371/journal.pone.0063201] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 04/03/2013] [Indexed: 11/19/2022] Open
Abstract
The use of recombinant viral vectors expressing T. gondii antigens is a safe and efficient approach to induce immune response against the parasite and a valuable tool for vaccine development. We have previously protected mice from toxoplasmosis by immunizing the animals with an adenovirus expressing the protein SAG1 (AdSAG1) of T. gondii. We are now looking for ways to improve the vaccination strategy and enhance protection. One limitation of homologous vaccinations (sequential doses of the same vector) is induction of anti-vector immune response that blocks cell transduction, restricts transgene expression and, consequently, compromises the overall outcome of vaccination. One way to avert the effects of anti-vector response is to use different viruses in prime and boost (heterologous vaccination). Bearing this in mind, we generated a modified Vaccinia Virus Ankara encoding SAG1 (MVASAG1), to be tested as boost agent after prime with AdSAG1. Although minor differences were observed in the magnitude of the anti-SAG1 immune response induced by each vaccination protocol, the heterologous immunization with AdSAG1 followed by MVASAG1 resulted in improved capacity to control brain cyst formation in a model of chronic toxoplasmosis in C57BL/6 mice.
Collapse
Affiliation(s)
- Érica Araújo Mendes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Flavio G. Fonseca
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Bárbara M. Casério
- Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Janaína P. Colina
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ricardo Tostes Gazzinelli
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
- Division of Infectious Disease and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail: (RTG); (BCC)
| | - Braulia C. Caetano
- Division of Infectious Disease and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail: (RTG); (BCC)
| |
Collapse
|
19
|
Alejo DM, Moraes MP, Liao X, Dias CC, Tulman ER, Diaz-San Segundo F, Rood D, Grubman MJ, Silbart LK. An adenovirus vectored mucosal adjuvant augments protection of mice immunized intranasally with an adenovirus-vectored foot-and-mouth disease virus subunit vaccine. Vaccine 2013; 31:2302-9. [PMID: 23499593 DOI: 10.1016/j.vaccine.2013.02.060] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 02/08/2013] [Accepted: 02/27/2013] [Indexed: 11/30/2022]
Abstract
Foot-and-mouth disease virus (FMDV) is a highly contagious pathogen that causes severe morbidity and economic losses to the livestock industry in many countries. The oral and respiratory mucosae are the main ports of entry of FMDV, so the stimulation of local immunity in these tissues may help prevent initial infection and viral spread. E. coli heat-labile enterotoxin (LT) has been described as one of the few molecules that have adjuvant activity at mucosal surfaces. The objective of this study was to evaluate the efficacy of replication-defective adenovirus 5 (Ad5) vectors encoding either of two LT-based mucosal adjuvants, LTB or LTR72. These vectored adjuvants were delivered intranasally to mice concurrent with an Ad5-FMDV vaccine (Ad5-A24) to assess their ability to augment mucosal and systemic humoral immune responses to Ad5-A24 and protection against FMDV. Mice receiving Ad5-A24 plus Ad5-LTR72 had higher levels of mucosal and systemic neutralizing antibodies than those receiving Ad5-A24 alone or Ad5-A24 plus Ad5-LTB. The vaccine plus Ad5-LTR72 group also demonstrated 100% survival after intradermal challenge with a lethal dose of homologous FMDV serotype A24. These results suggest that Ad5-LTR72 could be used as an important tool to enhance mucosal and systemic immunity against FMDV and potentially other pathogens with a common route of entry.
Collapse
Affiliation(s)
- Diana M Alejo
- Department of Animal Science, University of Connecticut, Storrs, CT 06269, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Yang J, Zhong M, Zhang Y, Zhang E, Sun Y, Cao Y, Li Y, Zhou D, He B, Chen Y, Yang Y, Yu J, Yan H. Antigen replacement of domains D2 and D3 in flagellin promotes mucosal IgA production and attenuates flagellin-induced inflammatory response after intranasal immunization. Hum Vaccin Immunother 2013; 9:1084-92. [PMID: 23377752 DOI: 10.4161/hv.23809] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Targeting early infection in mucosal sites is one of the primary goals for mucosal vaccines so as to prevent pathogen mucosal transmission and infection. The TLR5 agonist flagellin was deemed to be a mucosal adjuvant candidate for clinical usage. However, the high antigenicity of flagellin and the possible inflammatory injury induced by flagellin might restrict its clinical usage. Here HIV-1 p24 protein was selected as an antigen model and we replaced the main antigenicity region domains D2 and D3 of non-pathogenic E.coli-derived flagellin (KF). The derived soluble protein KFD-p24 3D was then compared with KF-p24, which fused p24 directly to the C-terminal of KF. In vitro and ex vivo experiments showed that KFD-p24 3D has lower TLR5 agonist efficacy and less immunocyte-activating efficacy. Interestingly, the production of KF- specific antibody was highly reduced, and KFD-p24 3D induced IgA-biased antibody responses in mucosal sites. Moreover, KFD-p24 3D induced far fewer systemic inflammatory responses and abrogated detectable inflammatory side effects on mice, even at the high dose. The properties of enhanced IgA generation and attenuated inflammatory responses broaden the safe-dose range of KFD-p24 3D flagellin, creating a potentially promising mucosal adjuvant.
Collapse
Affiliation(s)
- Jingyi Yang
- Mucosal Immunity Research Group; State Key Laboratory of Virology; Wuhan Institute of Virology; Chinese Academy of Sciences; Wuhan, P.R. China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Dziadek B, Brzostek A. Recombinant ROP2, ROP4, GRA4 and SAG1 antigen-cocktails as possible tools for immunoprophylaxis of toxoplasmosis: what's next? Bioengineered 2012; 3:358-64. [PMID: 22892593 DOI: 10.4161/bioe.21541] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Toxoplasmosis is a globally distributed foodborne zoonosis caused by a protozoan parasite Toxoplasma gondii. Usually asymptomatic in immunocompetent humans, toxoplasmosis is a serious clinical and veterinary problem often leading to lethal damage in an infected host. In order to overcome the exceptionally strong clinical and socio-economic impact of Toxoplasma infection, the construction of an effective vaccine inducing full immunoprotection against the parasite is an urgent issue. In the last two decades many live attenuated, subunit and DNA-based vaccines against toxoplasmosis have been studied, however only partial protection conferred by vaccination against chronic as well as acute infection has been achieved. Among various immunization strategies, no viable subunit vaccines based on recombinant secretory (ROP2, ROP4 and GRA4) and surface (SAG1) T. gondii proteins have been found as attractive tools for further studies. This is due to their high, but still partial, protective efficacy correlated with the induction of cellular and humoral immune responses.
Collapse
Affiliation(s)
- Bozena Dziadek
- Department of Immunoparasitology; University of Lodz, Lodz, Poland.
| | | |
Collapse
|
22
|
Zulpo DL, Headley SA, Biazzono L, da Cunha IAL, Igarashi M, de Barros LD, Taroda A, Cardim ST, Bogado ALG, Navarro IT, Garcia JL. Oocyst shedding in cats vaccinated by the nasal and rectal routes with crude rhoptry proteins of Toxoplasma gondii. Exp Parasitol 2012; 131:223-230. [PMID: 22542988 DOI: 10.1016/j.exppara.2012.04.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 03/30/2012] [Accepted: 04/09/2012] [Indexed: 10/28/2022]
Abstract
During this study, cats were immunized by the intranasal and rectal routes with crude rhoptry proteins of Toxoplasma gondii admixed with Quil-A. Twenty-five domestic short hair cats divided into five groups (n=5) were used during this evaluation: G1 and G3 cats received 200 μg of the rhoptry proteins with Quil-A (20 μg) by the intranasal and rectal routes, respectively; G2 and G4 cats received bovine serum albumin (BSA, 200 μg/dose) with Quil-A (20 μg); and G5 animals served as unvaccinated controls. All treatments were performed at days 0, 21, 42, and 63. The challenge was done with 800 cysts of the ME49 of T. gondii strain at day 70 (challenge day). The serum IgG, IgM, IgA, and fecal IgA antibody levels were evaluated by using the indirect enzyme-linked immunosorbent assay (ELISA). Some animals produced antibody levels beyond cut-off; however, two animals from G1 (OD(mean)=0.308, OD(cut-off)=0.200) and three from G3 (OD(mean)=0.254) demonstrated IgG levels on being challenged, with similar results occurring in two cats from G1 to IgM (OD(mean)=0.279, OD(cut-off)=0.200). Fecal IgA levels were detected in all G1 cats (OD(mean)=0.330, OD(cut-off)=0.065), and in one cat from G3 (OD(mean)=0.167). The serum and fecal humoral immune responses did not correlate with oocyst shedding. Oocyst shedding varied from 98.4% (G1), 87.5% (G2), 53.0% (G3), to 58% (G4), and was lower than that of G5 cats. The prepatent period of cats vaccinated intranasally (G1) was reduced from 6-9.6 to 2.8 days, suggesting protection of environmental contamination, considering cats as the primary source of contamination. The intranasally and rectally administered rhoptry vaccines were able to partially protect cats against T. gondii cysts on being challenged; however, the intranasal method of vaccination yielded better results relative to the rectal route.
Collapse
Affiliation(s)
- Dauton Luiz Zulpo
- Protozoology Laboratory, Departamento de Medicina Veterinária Preventiva, Universidade de Londrina-UEL, Londrina, PR, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Khosroshahi KH, Ghaffarifar F, Sharifi Z, D'Souza S, Dalimi A, Hassan ZM, Khoshzaban F. Comparing the effect of IL-12 genetic adjuvant and alum non-genetic adjuvant on the efficiency of the cocktail DNA vaccine containing plasmids encoding SAG-1 and ROP-2 of Toxoplasma gondii. Parasitol Res 2012; 111:403-11. [PMID: 22350714 DOI: 10.1007/s00436-012-2852-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2010] [Accepted: 02/02/2012] [Indexed: 11/28/2022]
Abstract
Various methods are available for enhancing the potency of DNA vaccines, including employment of different forms of adjuvant. The current study was carried out to evaluate and compare the effects of genetic and non-genetic adjuvants on the immune response stimulated by DNA vaccine. Thus, two adjuvants, IL-12 (genetic adjuvant) and aluminum hydroxide (alum, non-genetic adjuvant), were used with cocktail DNA vaccine containing plasmids encoding complete rhoptry antigen 2 (ROP-2) and surface major antigen 1 (SAG-1) of Toxoplasma gondii. The efficacy of pcROP2+pcSAG1 in stimulation of the immune response against toxoplasmosis with and without adjuvant was evaluated in female BALB/c mice by measuring the level of total IgG antibody and cytokines. The results obtained indicated that after challenging the mice with the fatal RH strain of T. gondii, the survival rates of mice immunized with pcROP2+pcSAG1 (DNA cocktail), pcSAG1+pcROP2+alum, and pcSAG1+pcROP2+IL-12 were significantly greater than that of the control groups (p<0.05). Moreover, measurement of total IgG antibody indicated the significant difference between the control and experimental groups (p<0.05). Finally, the results obtained by measurement of cytokines (IFN-γ and IL-4) showed high levels of IFN-γ and low levels of IL-4 in groups vaccinated with pcROP2+pcSAG1 (DNA cocktail), pcSAG1+pcROP2+alum, and pcSAG1+pcROP2+IL-12 as the experiment groups, in comparison with the controls groups (PBS, pc-DNA3, alum+PBS, and pCAGGS-IL-12+pcDNA3). The results of the study showed that use of adjuvants (IL-12 and alum) coincident with DNA cocktail leads to significant change in the survival rates of the experiment groups in comparison with control groups. Also, there is no significant difference between adjuvants to induce immune responses.
Collapse
|
24
|
Rashid I, Hedhli D, Moiré N, Pierre J, Debierre-Grockiego F, Dimier-Poisson I, Mévélec MN. Immunological responses induced by a DNA vaccine expressing RON4 and by immunogenic recombinant protein RON4 failed to protect mice against chronic toxoplasmosis. Vaccine 2011; 29:8838-46. [PMID: 21983362 DOI: 10.1016/j.vaccine.2011.09.099] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 09/20/2011] [Accepted: 09/25/2011] [Indexed: 11/15/2022]
Abstract
The development of an effective vaccine against Toxoplasma gondii infection is an important issue due to the seriousness of the related public health problems, and the economic importance of this parasitic disease worldwide. Rhoptry neck proteins (RONs) are components of the moving junction macromolecular complex formed during invasion. The aim of this study was to evaluate the vaccine potential of RON4 using two vaccination strategies: DNA vaccination by the intramuscular route, and recombinant protein vaccination by the nasal route. We produced recombinant RON4 protein (RON4S2) using the Schneider insect cells expression system, and validated its antigenicity and immunogenicity. We also constructed optimized plasmids encoding full length RON4 (pRON4), or only the N-terminal (pNRON4), or the C-terminal part (pCRON4) of RON4. CBA/J mice immunized with pRON4, pNRON4 or pCRON4 plus a plasmid encoding the granulocyte-macrophage-colony-stimulating factor showed high IgG titers against rRON4S2. Mice immunized by the nasal route with rRON4S2 plus cholera toxin exhibited low levels of anti-RON4S2 IgG antibodies, and no intestinal IgA antibodies specific to RON4 were detected. Both DNA and protein vaccination generated a mixed Th1/Th2 response polarized towards the IgG1 antibody isotype. Both DNA and protein vaccination primed CD4+ T cells in vivo. In addition to the production of IFN-γ, and IL-2, Il-10 and IL-5 were also produced by the spleen cells of the immunized mice stimulated with RON4S2, suggesting that a mixed Th1/Th2 type immune response occurred in all the immunized groups. No cytokine was detectable in stimulated mesenteric lymph nodes from mice immunized by the nasal route. Immune responses were induced by both DNA and protein vaccination, but failed to protect the mice against a subsequent oral challenge with T. gondii cysts. In conclusion, strategies designed to enhance the immunogenicity and to redirect the cellular response towards a Th1 type response against RON4 could lead to more encouraging results.
Collapse
Affiliation(s)
- Imran Rashid
- Université François Rabelais, INRA, UMR 0483 Université-INRA d'Immunologie Parasitaire, Vaccinologie et Biothérapie anti-infectieuse, IFR136 Agents Transmissibles et Infectiologie, UFR des Sciences Pharmaceutiques, 31 Avenue Monge, 37200 Tours, France
| | | | | | | | | | | | | |
Collapse
|
25
|
Igarashi M, Zulpo DL, Cunha IALD, Barros LD, Pereira VF, Taroda A, Navarro IT, Vidotto O, Vidotto MC, Jenkins MC, Garcia JL. Toxoplasma gondii: humoral and cellular immune response of BALB/c mice immunized via intranasal route with rTgROP2. ACTA ACUST UNITED AC 2011; 19:210-6. [PMID: 21184696 DOI: 10.1590/s1984-29612010000400004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 11/05/2010] [Indexed: 11/22/2022]
Abstract
TgROP2 is an intracellular protein associated with rhoptries of Toxoplama gondii and an antigen component of a candidate vaccine for toxoplasmosis. The purpose of the present study was to evaluate the efficacy of rTgROP2 to stimulate humoral and cellular immune responses in BALB/c mice via intranasal injection. TgROP2 partial coding sequence was (196-561) amplified by PCR from genomic T. gondii RH strain DNA and cloned into the pTrcHis expression vector. Escherichia coli Rosetta 2 cells transformed with pTrcHis-TgROP2 showed high levels (~1 mg.mL(-1)) of recombinant protein after 4 hours of IPTG induction. Recombinant TgROP2 exhibited an apparent Mr equal to 54 kDa. In order to test immunogenicity of the recombinant protein, 10 BALB/c mice received 10 µg of rROP2 protein + 10 µg of Quil-A via intranasal injection. Doses were administered at days 0, 21, and 42. Three animals were euthanized and used to evaluate cellular immune response on day 62. Five (50%) and two (20%) out of ten animals produced IgG (DO mean = 0.307; cut-off = 0.240) and IgA (DO mean = 0.133, cut-off = 0.101), respectively, by ELISA on day 62. The proliferation of splenocytes revealed high stimulation index (SI) when co-cultured with 5, 10 and 15 µg.mL(-1) of rTgROP2. These results indicate that intranasal immunization with recombinant protein ROP2 plus Quil-A can elicit both cellular and humoral immune responses in BALB/c mice.
Collapse
Affiliation(s)
- Michelle Igarashi
- Laboratório de Protozoologia, Departamento de Medicina Veterinária Preventiva, Universidade de Londrina, Londrina-PR, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Mendes EA, Caetano BC, Penido MLO, Bruna-Romero O, Gazzinelli RT. MyD88-dependent protective immunity elicited by adenovirus 5 expressing the surface antigen 1 from Toxoplasma gondii is mediated by CD8(+) T lymphocytes. Vaccine 2011; 29:4476-84. [PMID: 21549794 DOI: 10.1016/j.vaccine.2011.04.044] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 04/07/2011] [Accepted: 04/15/2011] [Indexed: 10/18/2022]
Abstract
Toxoplasma gondii is an intracellular parasite widely spread around the world. The surface antigens (SAG) 1, 2 and 3 are the main proteins expressed on the surface of T. gondii tachyzoites. Replication-defective adenovirus serotype 5 (rAd5) is one of the most potent recombinant viral vectors for eliciting T cell-mediated immunity in mice and humans. Here we show that vaccination with rAd5 expressing SAG1 (AdSAG1), but neither SAG2 nor SAG3, induces protective immunity in the highly susceptible C57BL/6 mice challenged with T. gondii. Furthermore, we evaluated different immunological components involved on viral induced protective immunity. We observed that host protection elicited by AdSAG1 is highly dependent on IL-12, IFN-γ and CD8(+) T lymphocytes. Importantly, the induction of protective immunity (T cell-derived IFN-γ) was also dependent on Myeloid Differentiation Factor 88 (MyD88), and thus, likely to involve Toll-like Receptors. We conclude that protective parasite specific-CD8(+) T cells are elicited by a mechanism that involves MyD88-dependent induction of IL-12.
Collapse
Affiliation(s)
- Erica A Mendes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | | | | | | | | |
Collapse
|
27
|
Humoral responses and immune protection in mice immunized with irradiated T. gondii tachyzoites and challenged with three genetically distinct strains of T. gondii. Immunol Lett 2011; 138:187-96. [PMID: 21545808 DOI: 10.1016/j.imlet.2011.04.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 04/08/2011] [Accepted: 04/18/2011] [Indexed: 11/21/2022]
Abstract
Toxoplasma gondii is an obligate intracellular parasite that infects a variety of mammals and birds. T. gondii also causes human toxoplasmosis; although toxoplasmosis is generally a benign disease, ocular, congenital or reactivated disease is associated with high numbers of disabled people. Infection occurs orally through the ingestion of meat containing cysts or by the intake of food or water contaminated with oocysts. Although the immune system responds to acute infection and mediates the clearance of tachyzoites, parasite cysts persist for the lifetime of the host in tissues such as the eye, muscle, and CNS. However, T. gondii RH strain tachyzoites irradiated with 255Gy do not cause residual infection and induce the same immunity as a natural infection. To assess the humoral response in BALB/c and C57BL/6J mice immunized with irradiated tachyzoites either by oral gavage (p.o.) or intraperitoneal (i.p.) injection, we analyzed total and high-affinity IgG and IgA antibodies in the serum. High levels of antigen-specific IgG were detected in the serum of parenterally immunized mice, with lower levels in mice immunized via the oral route. However, most serum antibodies exhibited low affinity for antigen in both mice strain. We also found antigen specific IgA antibodies in the stools of the mice, especially in orally immunized BALB/c mice. Examination of bone marrow and spleen cells demonstrated that both groups of immunized mice clearly produced specific IgG, at levels comparable to chronic infection, suggesting the generation of IgG specific memory. Next, we challenged i.p. or p.o. immunized mice with cysts from ME49, VEG or P strains of T. gondii. Oral immunization resulted in partial protection as compared to challenged naive mice; these findings were more evident in highly pathogenic ME49 strain challenge. Additionally, we found that while mucosal IgA was important for protection against infection, antigen-specific IgG antibodies were involved with protection against disease and disease pathogenesis. Most antigen responsive cells in culture produced specific high-affinity IgG after immunization, diverse of the findings in serum IgG or from cells after infection, which produced low proportion of high-avidity IgG.
Collapse
|
28
|
da Hora VP, Conceição FR, Dellagostin OA, Doolan DL. Non-toxic derivatives of LT as potent adjuvants. Vaccine 2011; 29:1538-44. [DOI: 10.1016/j.vaccine.2010.11.091] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Revised: 11/10/2010] [Accepted: 11/30/2010] [Indexed: 12/21/2022]
|
29
|
Evaluation of three recombinant multi-antigenic vaccines composed of surface and secretory antigens of Toxoplasma gondii in murine models of experimental toxoplasmosis. Vaccine 2011; 29:821-30. [DOI: 10.1016/j.vaccine.2010.11.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Revised: 07/16/2010] [Accepted: 11/02/2010] [Indexed: 11/24/2022]
|
30
|
Jongert E, Roberts CW, Gargano N, Förster-Waldl E, Förster-Wald E, Petersen E. Vaccines against Toxoplasma gondii: challenges and opportunities. Mem Inst Oswaldo Cruz 2010; 104:252-66. [PMID: 19430651 DOI: 10.1590/s0074-02762009000200019] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2008] [Accepted: 12/04/2008] [Indexed: 12/21/2022] Open
Abstract
Development of vaccines against Toxoplasma gondii infection in humans is of high priority, given the high burden of disease in some areas of the world like South America, and the lack of effective drugs with few adverse effects. Rodent models have been used in research on vaccines against T. gondii over the past decades. However, regardless of the vaccine construct, the vaccines have not been able to induce protective immunity when the organism is challenged with T. gondii, either directly or via a vector. Only a few live, attenuated T. gondii strains used for immunization have been able to confer protective immunity, which is measured by a lack of tissue cysts after challenge. Furthermore, challenge with low virulence strains, especially strains with genotype II, will probably be insufficient to provide protection against the more virulent T. gondii strains, such as those with genotypes I or II, or those genotypes from South America not belonging to genotype I, II or III. Future studies should use animal models besides rodents, and challenges should be performed with at least one genotype II T. gondii and one of the more virulent genotypes. Endpoints like maternal-foetal transmission and prevention of eye disease are important in addition to the traditional endpoint of survival or reduction in numbers of brain cysts after challenge.
Collapse
Affiliation(s)
- Erik Jongert
- Laboratory for Toxoplasmosis, Pasteur Institute of Brussels, Scientific Institute for Public Health, Brussels, Belgium
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
Mucosal vaccination offers the advantage of blocking pathogens at the portal of entry, improving patient's compliance, facilitating vaccine delivery, and decreasing the risk of unwanted spread of infectious agents via contaminated syringes.Recent advances in vaccinology have created an array of vaccine constructs that can be delivered to mucosal surfaces of the respiratory, gastrointestinal, and genitourinary tracts using intranasal, oral, and vaginal routes. Due to the different characteristics of mucosal immune response, as compared with systemic response, mucosal immunization requires particular methods of antigen presentation. Well-tolerated adjuvants that enhance the efficacy of such vaccines will play an important role in mucosal immunization. Among promising mucosal adjuvants, mutants of cholera toxin and the closely related heat-labile enterotoxin (LT) of enterotoxigenic Escherichia coli present powerful tools, augmenting the local and systemic serum antibody response to co-administered antigens.In this chapter, we describe the formulation and application of vaccines using the genetically modified LTK63 mutant as a prototype of the family of these mucosal adjuvants and the tools to determine its activity in the mouse model.
Collapse
|
32
|
Intraperitoneal and intra-nasal vaccination of mice with three distinct recombinant Neospora caninum antigens results in differential effects with regard to protection against experimental challenge with Neospora caninum tachyzoites. Parasitology 2009; 137:229-40. [PMID: 19835644 DOI: 10.1017/s0031182009991259] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Recombinant NcPDI(recNcPDI), NcROP2(recNcROP2), and NcMAG1(recNcMAG1) were expressed in Escherichia coli and purified, and evaluated as potential vaccine candidates by employing the C57Bl/6 mouse cerebral infection model. Intraperitoneal application of these proteins suspended in saponin adjuvants lead to protection against disease in 50% and 70% of mice vaccinated with recNcMAG1 and recNcROP2, respectively, while only 20% of mice vaccinated with recNcPDI remained without clinical signs. In contrast, a 90% protection rate was achieved following intra-nasal vaccination with recNcPDI emulsified in cholera toxin. Only 1 mouse vaccinated intra-nasally with recNcMAG1 survived the challenge infection, and protection achieved with intra-nasally applied recNcROP2 was at 60%. Determination of cerebral parasite burdens by real-time PCR showed that these were significantly reduced only in recNcROP2-vaccinated animals (following intraperitoneal and intra-nasal application) and in recNcPDI-vaccinated mice (intra-nasal application only). Quantification of viable tachyzoites in brain tissue of intra-nasally vaccinated mice showed that immunization with recNcPDI resulted in significantly decreased numbers of live parasites. These data show that, besides the nature of the antigen, the protective effect of vaccination also depends largely on the route of antigen delivery. In the case of recNcPDI, the intra-nasal route provides a platform to generate a highly protective immune response.
Collapse
|
33
|
Wang H, He S, Yao Y, Cong H, Zhao H, Li T, Zhu XQ. Toxoplasma gondii: Protective effect of an intranasal SAG1 and MIC4 DNA vaccine in mice. Exp Parasitol 2009; 122:226-32. [DOI: 10.1016/j.exppara.2009.04.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2009] [Revised: 02/19/2009] [Accepted: 04/04/2009] [Indexed: 10/20/2022]
|
34
|
Hedhli D, Dimier-Poisson I, Judge JW, Rosenberg B, Mévélec MN. Protective immunity against Toxoplasma challenge in mice by coadministration of T. gondii antigens and Eimeria profilin-like protein as an adjuvant. Vaccine 2009; 27:2274-81. [DOI: 10.1016/j.vaccine.2009.01.100] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Revised: 01/15/2009] [Accepted: 01/22/2009] [Indexed: 01/01/2023]
|
35
|
Abstract
Toxoplasma gondii is a parasite that infects animals and humans worldwide. Despite the current knowledge of immunology, pathology and genetics related to the parasite, a safe vaccine for prevention of the infection in both humans and animals does not exist. Here, we review some aspects concerning vaccination against T. gondii.
Collapse
Affiliation(s)
- João Luis Garcia
- Department of Preventive Veterinary Medicine, Londrina State University, Campus Universitário, Rodovia Celso Garcia Cid, Pr 445 Km 380, Cx. Postal 6001, Londrina, PR 86051-990, Brazil.
| |
Collapse
|
36
|
Jongert E, Verhelst D, Abady M, Petersen E, Gargano N. Protective Th1 immune responses against chronic toxoplasmosis induced by a protein–protein vaccine combination but not by its DNA–protein counterpart. Vaccine 2008; 26:5289-95. [DOI: 10.1016/j.vaccine.2008.07.032] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2008] [Revised: 07/08/2008] [Accepted: 07/14/2008] [Indexed: 11/26/2022]
|
37
|
Toxoplasma gondii: Evaluation of an intranasal vaccine using recombinant proteins against brain cyst formation in BALB/c mice. Exp Parasitol 2008; 118:386-92. [DOI: 10.1016/j.exppara.2007.10.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2007] [Revised: 09/05/2007] [Accepted: 10/02/2007] [Indexed: 11/19/2022]
|
38
|
Beauvillain C, Ruiz S, Guiton R, Bout D, Dimier-Poisson I. A vaccine based on exosomes secreted by a dendritic cell line confers protection against T. gondii infection in syngeneic and allogeneic mice. Microbes Infect 2007; 9:1614-22. [PMID: 17905628 DOI: 10.1016/j.micinf.2007.07.002] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2006] [Revised: 06/29/2007] [Accepted: 07/07/2007] [Indexed: 01/05/2023]
Abstract
Our results show that exosomes secreted by SRDC pulsed in vitro with Toxoplasma gondii-derived antigens (Exo-TAg) induced protective responses against infection with the parasite in both syngeneic and allogeneic mice. After oral infection, syngeneic CBA/J mice exhibited significantly fewer cysts in their brains and allogeneic C57BL/6 mice survived. This protection was associated with strong humoral responses in vivo in serum from both CBA/J and C57BL/6 mice, and with high levels of anti-TAg IgA antibodies in intestinal secretions from CBA/J mice alone. Furthermore, strong cellular responses in vivo were observed in both mouse models. Cellular proliferation was associated with cytokines production by spleen and mesenteric lymph node cells. The results presented here show that exosomes are nucleic acid free vesicles that are able to induce immune responses correlated with protection against parasitic infections in both syngeneic and allogeneic mice. They could constitute an efficient tool for use in vaccination and antitumor strategies based on exosomes.
Collapse
Affiliation(s)
- Céline Beauvillain
- Université François-Rabelais, INRA, UMR 0483 Université-INRA d'Immunologie Parasitaire et Vaccinologie, IFR des Agents Transmissibles et Infectiologie, UFR des Sciences Pharmaceutiques, Tours, France.
| | | | | | | | | |
Collapse
|
39
|
Tritto E, Muzzi A, Pesce I, Monaci E, Nuti S, Galli G, Wack A, Rappuoli R, Hussell T, De Gregorio E. The Acquired Immune Response to the Mucosal Adjuvant LTK63 Imprints the Mouse Lung with a Protective Signature. THE JOURNAL OF IMMUNOLOGY 2007; 179:5346-57. [PMID: 17911621 DOI: 10.4049/jimmunol.179.8.5346] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
LTK63, a nontoxic mutant of Escherichia coli heat labile enterotoxin (LT), is a potent and safe mucosal adjuvant that has also been shown to confer generic protection to several respiratory pathogens. To understand the mechanisms of action underlying the LTK63 protective effect, we analyzed the molecular and cellular events triggered by its administration in vivo. We show here that LTK63 intrapulmonary administration induced in the mouse lung a specific gene expression signature characterized by the up-regulation of cell cycle genes, several host defense genes, chemokines, chemokine receptors, and immune cell-associated genes. Such a transcriptional profile reflected the activation of alveolar macrophages and the recruitment to the lung of T and B cells and innate immune cells such as granulocytes, NK, and dendritic cells. All of these events were T cell dependent and specific for LTK63 because they were absent in SCID and nude mice. Additionally, we showed that LTK63 induces a potent adaptive immune response against itself directed to the lung. We propose that acquired response to LTK63 is the driving force for the local recruitment of both adaptive and innate immune cells. Our data suggest that LTK63 acts as an airway infection mimic that establishes a generic protective environment limiting respiratory infection by innate immune mechanisms and by improving adaptive responses to invading pathogens.
Collapse
|
40
|
Garcia JL, Navarro IT, Biazzono L, Freire RL, da Silva Guimarães Junior J, Cryssafidis AL, Bugni FM, da Cunha IAL, Hamada FN, Dias RCF. Protective activity against oocyst shedding in cats vaccinated with crude rhoptry proteins of the Toxoplasma gondii by the intranasal route. Vet Parasitol 2007; 145:197-206. [PMID: 17296268 DOI: 10.1016/j.vetpar.2007.01.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2006] [Revised: 01/08/2007] [Accepted: 01/15/2007] [Indexed: 11/26/2022]
Abstract
This study evaluated a vaccine made from crude rhoptry proteins of Toxoplasma gondii with Quil-A, which was administered to cats by the intranasal route. Eleven short-hair domestic cats were divided into four groups: G1 (n=3) received three doses (200 microg/dose) of the rhoptry vaccine with Quil-A (20 microg); G2 (n=3) received PBS with Quil-A (20 microg); G3 (n=3) and G4 (n=2) received only PBS. Treatments were administered at days 0, 21, and 42 by the intranasal route. Challenge was done to G1, G2, and G3 animals with 600 cysts of the VEG strain on day 51 (challenge day); G4 animals were unchallenged. The anti-T. gondii IgG and IgA antibody levels from sera were measured by indirect enzyme-linked immunosorbent assay (ELISA). At challenge, two animals from G1 revealed antibody levels for both IgG and IgA; oocysts were not detected in feces of these two cats. There were no differences in hematological values between groups throughout the experiment (p>0.10). Preventable fractions were 67% in G1 and 0% in G2 and G3. Comparatively, G1 animals shed 89.3% and 90.8% less oocysts than G3 and G4, respectively. Two out of three cats were protected against T. gondii oocyst shedding when the rhoptry vaccine was administered by the intranasal route. This is the first study using crude rhoptry proteins as vaccine by the intranasal route in cats to evaluate protection against oocysts shedding.
Collapse
Affiliation(s)
- João Luis Garcia
- Protozoology laboratory, Departamento de Medicina Veterinária Preventiva, Universidade Estadual de Londrina, Postal Box 6001, 86050-970 Londrina, PR, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Teixeira L, Botelho AS, Batista AR, Meireles CS, Ribeiro A, Domingues HS, Correia Da Costa JM, Castro AG, Faustino AMR, Vilanova M. Analysis of the immune response to Neospora caninum in a model of intragastric infection in mice. Parasite Immunol 2007; 29:23-36. [PMID: 17187652 DOI: 10.1111/j.1365-3024.2006.00911.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
To study experimental Neospora caninum infection initiated at the gastrointestinal tract, Toll-like Receptor 4- and functional IL-12Rbeta2 chain-deficient C57BL/10 ScCr mice were challenged intragastrically with 5 x 10(6) N. caninum tachyzoites. All parasite-inoculated mice eventually died with disseminated infection. In contrast, immunocompetent BALB/c mice challenged with 1 x 10(7) N. caninum tachyzoites by the intragastric (i.g.) or the intraperitoneal (i.p.) route remained alive for at least 6 months. Expansion of splenic B- and T-cells, the latter displaying both activated and regulatory phenotypes, and increased levels of IFN-gamma and IL-10 mRNA were detected in both groups of infected BALB/c mice compared with non-infected controls, whereas in the Peyer's patches only IFN-gamma mRNA levels were found to be increased. Parasite-specific IgG1, IgG2a and IgA antibody levels were elevated in the sera of all infected mice, whereas increased N. caninum-specific IgA levels were detected in intestinal lavage fluids of i.g. challenged mice only. These results show that N. caninum infection can be successfully established in mice by i.g. administration of tachyzoites. They also show that the immune response elicited in i.g. or i.p. infected BALB/c mice, although conferring some degree of protection, was not sufficient for complete parasite clearance.
Collapse
Affiliation(s)
- L Teixeira
- Instituto de Ciências Biomédicas de Abel Salazar, Largo do Professor Abel Salazar 2, 4099-003, Porto, Portugal
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Döşkaya M, Kalantari-Dehaghi M, Walsh CM, Hiszczyńska-Sawicka E, Davies DH, Felgner PL, Larsen LSZ, Lathrop RH, Hatfield GW, Schulz JR, Gürüz Y, Jurnak F. GRA1 protein vaccine confers better immune response compared to codon-optimized GRA1 DNA vaccine. Vaccine 2007; 25:1824-37. [PMID: 17234306 DOI: 10.1016/j.vaccine.2006.10.060] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2006] [Revised: 10/26/2006] [Accepted: 10/30/2006] [Indexed: 01/15/2023]
Abstract
The present study evaluates immunogenicity and protection potency of a codon-optimized GRA1 DNA vaccine, wild type GRA1 DNA vaccine and an adjuvanted recombinant GRA1 protein vaccine candidate in BALB/c mice against lethal toxoplasmosis. Of the three GRA1 vaccines tested, the recombinant GRA1 protein vaccine results reveal significant increase in immune response and prolonged survival against acute toxoplasmosis compared to DNA vaccinations. Immune response and protection conferred by codon-optimized GRA1 DNA vaccine was slightly better than wild type GRA1 DNA vaccine.
Collapse
Affiliation(s)
- Mert Döşkaya
- Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Dietrich J, Andersen C, Rappuoli R, Doherty TM, Jensen CG, Andersen P. Mucosal administration of Ag85B-ESAT-6 protects against infection with Mycobacterium tuberculosis and boosts prior bacillus Calmette-Guerin immunity. THE JOURNAL OF IMMUNOLOGY 2006; 177:6353-60. [PMID: 17056566 DOI: 10.4049/jimmunol.177.9.6353] [Citation(s) in RCA: 147] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have examined the intranasal administration of a vaccine against Mycobacterium tuberculosis (M.tb) consisting of the mucosal adjuvant LTK63 and the Ag Ag85B-ESAT-6. Vaccination with LTK63/Ag85B-ESAT-6 gave a strong and sustained Th1 response mediated by IFN-gamma-secreting CD4 cells, which led to long-lasting protection against tuberculosis, equivalent to that observed with bacillus Calmette-Guérin (BCG) or Ag85B-ESAT-6 in dimethyldioctadecylammonium bromide/monophosphoryl lipid A. Because a crucial element of novel vaccine strategies is the ability to boost BCG-derived immunity, we also tested whether LTK63/Ag85B-ESAT-6 could act as a BCG booster vaccine in BCG-vaccinated mice. We found that vaccinating with LTK63/Ag85B-ESAT-6 strongly boosted prior BCG-stimulated immunity. Compared with BCG-vaccinated nonboosted mice, we observed that infection with M.tb led to a significant increase in anti-M.tb-specific CD4 T cells in the lungs of LTK63/Ag85B-ESAT-6-boosted animals. This correlated with a significant increase in the protection against M.tb in LTK63/Ag85B-ESAT-6-boosted mice, compared with BCG-vaccinated animals. Thus, LTK63/Ag85B-ESAT-6 represents an efficient preventive vaccine against tuberculosis with a strong ability to boost prior BCG immunity.
Collapse
Affiliation(s)
- Jes Dietrich
- Department of Infectious Disease Immunology, Statens Serum Institute, Artillerivej 5, DK-2300 Copenhagen, Denmark.
| | | | | | | | | | | |
Collapse
|
44
|
Gatkowska J, Hiszczynska-Sawicka E, Kur J, Holec L, Dlugonska H. Toxoplasma gondii: An evaluation of diagnostic value of recombinant antigens in a murine model. Exp Parasitol 2006; 114:220-7. [DOI: 10.1016/j.exppara.2006.03.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2006] [Revised: 03/15/2006] [Accepted: 03/25/2006] [Indexed: 11/16/2022]
|
45
|
Caetano BC, Bruña-Romero O, Fux B, Mendes EA, Penido MLO, Gazzinelli RT. Vaccination with replication-deficient recombinant adenoviruses encoding the main surface antigens of toxoplasma gondii induces immune response and protection against infection in mice. Hum Gene Ther 2006; 17:415-26. [PMID: 16610929 DOI: 10.1089/hum.2006.17.415] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We have generated recombinant adenoviruses encoding three genetically modified surface antigens (SAGs) of the parasite Toxoplasma gondii, that is, AdSAG1, AdSAG2, and AdSAG3. Modifications included the removal of their glycosylphosphatidylinositol (GPI) anchoring motifs and, in some cases, the exchange of the native signal peptide for influenza virus hemagglutinin signal sequence. Adenovirus immunization of BALB/c mice elicited potent antibody responses against each protein, displaying a significant bias toward a helper T cell type 1 (Th1) profile in animals vaccinated with AdSAG1. Furthermore, the presence of parasite-specific IFN-gamma-producing T cells was analyzed by proliferation assays and enzyme-linked immunospot assays in the same animals. Splenocytes from immunized mice secreted IFN-gamma after in vitro stimulation with tachyzoite lysate antigen or with a fraction enriched for membrane-purified GPI-anchored proteins (F3) from the T. gondii tachyzoite surface. Epitopes recognized by CD8+ T cells were identified in SAG1 and SAG3, but not SAG2, sequences, although this protein also induced a specific response. We also tested the capacity of the immune responses detected to protect mice against a challenge with live T. gondii parasites. Although no protection was observed against tachyzoites of the highly virulent RH strain, a significant reduction in cyst loads in the brain was observed in animals challenged with the P-Br strain. Thus, up to 80% of the cysts were eliminated from animals vaccinated with a mixture of the three recombinant viruses. Because adenoviruses seemed capable of inducing Th1-biased protective immune responses against T. gondii antigens, other parasite antigens should be tested alone or in combination with those described here to further develop a protective vaccine against toxoplasmosis.
Collapse
MESH Headings
- Adenoviridae
- Animals
- Antigens, Protozoan/genetics
- Antigens, Protozoan/immunology
- Female
- Gene Deletion
- Immunity, Active
- Immunity, Cellular
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Mice
- Mice, Inbred BALB C
- Protozoan Proteins/genetics
- Protozoan Proteins/immunology
- Protozoan Vaccines/genetics
- Protozoan Vaccines/immunology
- Recombination, Genetic
- Toxoplasma/immunology
- Toxoplasmosis, Animal/immunology
- Toxoplasmosis, Animal/prevention & control
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Virus Replication/genetics
Collapse
Affiliation(s)
- Bráulia C Caetano
- Laboratório de Imunoparasitologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | | | | | | | | | | |
Collapse
|
46
|
Caetano BC, Bruna-Romero O, Fux B, Mendes EA, Penido MLO, Gazzinelli RT. Vaccination with Replication-Deficient Recombinant Adenoviruses Encoding the Main Surface Antigens of Toxoplasma gondii Induces Immune Response and Protection Against Infection in Mice. Hum Gene Ther 2006. [DOI: 10.1089/hum.2006.17.ft-199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
47
|
Flori P, Tardy L, Jacquet A, Bellete B, Hafid J, Raberin H, Tran Manh Sung R. Effect of rSAG-1(P30) immunisation on the circulating and tissue parasites in guinea pigs as determined by quantitative PCR. Parasitol Res 2006; 98:511-8. [PMID: 16416293 DOI: 10.1007/s00436-005-0074-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2005] [Accepted: 10/26/2005] [Indexed: 01/22/2023]
Abstract
The efficacy of immunisation with Toxoplasma gondii recombinant protein (rSAG-1) was evaluated in the guinea pig model. For the infectious challenge, two strains, namely, strain C56 (10,000 tachyzoites) and strain 76K (100 cysts), were used to infect a group of 32 guinea pigs each. The circulating, cerebral and pulmonary parasite loads were determined with the real-time polymerase chain reaction (PCR) after immunisation. With the C56 strain, immunisation showed high activity with a reduction of greater than 1 log of the circulating and tissue parasite loads. Thus, there was a significantly lower circulating parasite load in the rSAG-1 + adjuvant group (0.5+/-1.5 Eq parasites/ml) as compared to that in the control group (67+/-110 Eq parasites/ml; p<0.05). In the same manner, the cerebral parasite load was much lower in the rSAG-1 + adjuvant group (10+/-20 Eq parasites/g) than that in the control group (339+/-291 Eq parasites/g; p<0.01). On the other hand, with the 76K strain, the effect of immunisation was much less and that only on the pulmonary parasite load [p(lung)<0.05]. This could be due to the use of different strains and stages of the parasite and/or the difference in the route of administration for challenge. The quantitative PCR technique used has shown a good correlation with animal inoculation, and when associated with the guinea pig model, it seems to be a useful and reproducible technique for future vaccine studies.
Collapse
Affiliation(s)
- Pierre Flori
- Laboratoire de Parasitologie-Mycologie, C.H.U. de Saint Etienne, Hôpital Nord, 42055, Saint-Etienne, cedex 2, France.
| | | | | | | | | | | | | |
Collapse
|
48
|
Howe DK, Gaji RY, Mroz-Barrett M, Gubbels MJ, Striepen B, Stamper S. Sarcocystis neurona merozoites express a family of immunogenic surface antigens that are orthologues of the Toxoplasma gondii surface antigens (SAGs) and SAG-related sequences. Infect Immun 2005; 73:1023-33. [PMID: 15664946 PMCID: PMC546929 DOI: 10.1128/iai.73.2.1023-1033.2005] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Sarcocystis neurona is a member of the Apicomplexa that causes myelitis and encephalitis in horses but normally cycles between the opossum and small mammals. Analysis of an S. neurona expressed sequence tag (EST) database revealed four paralogous proteins that exhibit clear homology to the family of surface antigens (SAGs) and SAG-related sequences of Toxoplasma gondii. The primary peptide sequences of the S. neurona proteins are consistent with the two-domain structure that has been described for the T. gondii SAGs, and each was predicted to have an amino-terminal signal peptide and a carboxyl-terminal glycolipid anchor addition site, suggesting surface localization. All four proteins were confirmed to be membrane associated and displayed on the surface of S. neurona merozoites. Due to their surface localization and homology to T. gondii surface antigens, these S. neurona proteins were designated SnSAG1, SnSAG2, SnSAG3, and SnSAG4. Consistent with their homology, the SnSAGs elicited a robust immune response in infected and immunized animals, and their conserved structure further suggests that the SnSAGs similarly serve as adhesins for attachment to host cells. Whether the S. neurona SAG family is as extensive as the T. gondii SAG family remains unresolved, but it is probable that additional SnSAGs will be revealed as more S. neurona ESTs are generated. The existence of an SnSAG family in S. neurona indicates that expression of multiple related surface antigens is not unique to the ubiquitous organism T. gondii. Instead, the SAG gene family is a common trait that presumably has an essential, conserved function(s).
Collapse
Affiliation(s)
- Daniel K Howe
- Department of Veterinary Science, 108 Gluck Equine Research Center, University of Kentucky, Lexington, KY 40546-0099, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Williams AE, Edwards L, Humphreys IR, Snelgrove R, Rae A, Rappuoli R, Hussell T. Innate imprinting by the modified heat-labile toxin of Escherichia coli (LTK63) provides generic protection against lung infectious disease. THE JOURNAL OF IMMUNOLOGY 2005; 173:7435-43. [PMID: 15585869 DOI: 10.4049/jimmunol.173.12.7435] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In a healthy individual, the lung contains few lymphoid cells. However, amplified immune responses, as exemplified during lung infection, can cause extensive tissue damage. We have previously demonstrated that one lung infection modulates the immunopathological outcome to a subsequent unrelated pathogen. Mimicking heterologous immunity may provide a means of enhancing both innate and acquired immunity. We now show that prior lung administration of a modified heat-labile toxin from Escherichia coli (LTK63) enhances immunity to respiratory syncytial virus, influenza virus, and the fungus Cryptococcus neoformans. Treatment with LTK63 decreased lung inflammation and tissue damage and improved the ability to resolve the infection. APCs expressing the activation markers MHC class II, CD80, and CD40 increased in number in the lung. LTK63 treatment increased the pathogen-specific IgA response in the nasal mucosa and simultaneously decreased inflammatory cytokine production (IFN-gamma and TNF-alpha) after infection. The number of activated CD8(+)CD44(+) T cells and the respiratory syncytial virus- or influenza-specific CD8-proliferative responses increased, although the total inflammatory infiltrate was reduced. LTK63 treatment matured lung APCs (LTK63 prevented efficient presentation of whole OVA to DO11.10 cells, whereas OVA peptide presentation was unaffected), enhanced immunity in both a Th1 and Th2 environment, was long lasting, and was not pathogen or host strain specific; the protective effects were partially independent of T and B cells. Innate imprinting by toxin-based immunotherapeutics may provide generic protection against infectious disease in the lung, without the need for coadministered pathogen-specific Ag.
Collapse
Affiliation(s)
- Andrew Evan Williams
- Centre For Molecular Microbiology and Infection, Department of Biological Sciences, Imperial College London, London, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
50
|
Hafid J, Vincent N, Flori P, Bellete B, Raberin H, Sung RTM. Production of antibodies in murine mucosal immunization with Toxoplasma gondii excreted/secreted antigens. Vet Parasitol 2004; 128:23-8. [PMID: 15725529 DOI: 10.1016/j.vetpar.2004.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2004] [Revised: 09/09/2004] [Accepted: 11/01/2004] [Indexed: 11/23/2022]
Abstract
Toxoplasmagondii RH strain excreted/secreted antigens (ESA) were administrated weekly by the oral route, to two groups of 40 OF1 mice for 4 weeks. One group received ESA associated with cholera toxin (CT+) and the other, ESA only (CT-). Five animals from each group were sacrificed from day 4 (D4) to D49 following the first immunization and their feces and sera were collected and tested by ELISA for IgA, IgG and IgM antibody detection. In feces, IgA antibodies were detected on D4 and on D12 in the CT+ and CT- groups, respectively, and they persisted up to D49. IgG antibodies were detected from D12 to D41 in the CT+ group and on D12 only in the CT- group. No IgM antibodies were detected. In sera, IgA antibodies were detected on D27, D41 and D49 only in the CT+ group. IgG and IgM antibodies were found on D12 and D4, respectively, in the CT+ group and starting from D27 in the CT- group. To our knowledge, this is the first demonstration that ESA, with or without CT, are immunogenic when administrated by the oral route.
Collapse
Affiliation(s)
- Jamal Hafid
- Groupe immunité des muqueuses et agents pathogènes (GIMAP), Faculté de médecine Jacques Lisfranc, 15 rue Ambroise Paré, 42023 Saint Etienne Cedex 02, France.
| | | | | | | | | | | |
Collapse
|