1
|
Somé GF, Séré M, Somda BM, Dayo GK, Ouédraogo GA, Boulangé A, Maarifi G, Chantal I, Berthier-Teyssedre D, Thévenon S. Immune Response in Cattle Trypanosomosis and Trypanotolerance: Main Findings and Gaps. Parasite Immunol 2024; 46:e13075. [PMID: 39508487 DOI: 10.1111/pim.13075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/30/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024]
Abstract
Trypanosome parasites of the genus Trypanosoma cause African animal trypanosomosis, a devastating livestock disease plaguing sub-Saharan Africa. Unlike many protozoan parasites, these extracellular blood-borne pathogens directly engage the host's immune system. While the mouse model has provided valuable insights, a comprehensive understanding of the bovine immune response to trypanosomes remains elusive. Addressing the immune response in cattle, the most relevant host species, and how it takes part in mitigating the negative impact of the disease could contribute to setting up sustainable control strategies. This review summarises the current knowledge of the immune response in cattle during trypanosomosis. Following a brief overview of infection processes and bovine trypanotolerance, we present advances in the regulation of host innate, inflammatory and adaptive responses and delve into the key immunological players involved in immunoactivities and immunosuppression. We discuss how these mechanisms contribute to tolerance or susceptibility to infection, highlighting critical gaps in knowledge that require further investigation.
Collapse
Affiliation(s)
- Gnohion Fabrice Somé
- Centre International de Recherche-Développement Sur l'Elevage en Zone Subhumide (CIRDES), Bobo-Dioulasso, Burkina Faso
- Cirad, UMR INTERTRYP, Montpellier, France
- INTERTRYP, Université de Montpellier, Cirad, IRD, Montpellier, France
| | - Modou Séré
- Centre International de Recherche-Développement Sur l'Elevage en Zone Subhumide (CIRDES), Bobo-Dioulasso, Burkina Faso
- Université Daniel-Ouezzin-COULIBALLY, Dédougou, Burkina Faso
| | - Bienvenu Martin Somda
- Centre International de Recherche-Développement Sur l'Elevage en Zone Subhumide (CIRDES), Bobo-Dioulasso, Burkina Faso
- Université Nazi BONI, Bobo-Dioulasso, Burkina Faso
| | - Guiguigbaza-Kossigan Dayo
- Centre International de Recherche-Développement Sur l'Elevage en Zone Subhumide (CIRDES), Bobo-Dioulasso, Burkina Faso
| | | | - Alain Boulangé
- Cirad, UMR INTERTRYP, Montpellier, France
- INTERTRYP, Université de Montpellier, Cirad, IRD, Montpellier, France
| | - Ghizlane Maarifi
- Cirad, UMR INTERTRYP, Montpellier, France
- INTERTRYP, Université de Montpellier, Cirad, IRD, Montpellier, France
| | - Isabelle Chantal
- Cirad, UMR INTERTRYP, Montpellier, France
- INTERTRYP, Université de Montpellier, Cirad, IRD, Montpellier, France
| | - David Berthier-Teyssedre
- Cirad, UMR INTERTRYP, Montpellier, France
- INTERTRYP, Université de Montpellier, Cirad, IRD, Montpellier, France
| | - Sophie Thévenon
- Cirad, UMR INTERTRYP, Montpellier, France
- INTERTRYP, Université de Montpellier, Cirad, IRD, Montpellier, France
| |
Collapse
|
2
|
Morrison LJ, Steketee PC, Tettey MD, Matthews KR. Pathogenicity and virulence of African trypanosomes: From laboratory models to clinically relevant hosts. Virulence 2023; 14:2150445. [PMID: 36419235 DOI: 10.1080/21505594.2022.2150445] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022] Open
Abstract
African trypanosomes are vector-borne protozoa, which cause significant human and animal disease across sub-Saharan Africa, and animal disease across Asia and South America. In humans, infection is caused by variants of Trypanosoma brucei, and is characterized by varying rate of progression to neurological disease, caused by parasites exiting the vasculature and entering the brain. Animal disease is caused by multiple species of trypanosome, primarily T. congolense, T. vivax, and T. brucei. These trypanosomes also infect multiple species of mammalian host, and this complexity of trypanosome and host diversity is reflected in the spectrum of severity of disease in animal trypanosomiasis, ranging from hyperacute infections associated with mortality to long-term chronic infections, and is also a main reason why designing interventions for animal trypanosomiasis is so challenging. In this review, we will provide an overview of the current understanding of trypanosome determinants of infection progression and severity, covering laboratory models of disease, as well as human and livestock disease. We will also highlight gaps in knowledge and capabilities, which represent opportunities to both further our fundamental understanding of how trypanosomes cause disease, as well as facilitating the development of the novel interventions that are so badly needed to reduce the burden of disease caused by these important pathogens.
Collapse
Affiliation(s)
- Liam J Morrison
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Pieter C Steketee
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Mabel D Tettey
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Keith R Matthews
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
3
|
Abstract
African trypanosomes are bloodstream protozoan parasites that infect mammals including humans, where they cause sleeping sickness. Long-lasting infection is required to favor parasite transmission between hosts. Therefore, trypanosomes have developed strategies to continuously escape innate and adaptive responses of the immune system, while also preventing premature death of the host. The pathology linked to infection mainly results from inflammation and includes anemia and brain dysfunction in addition to loss of specificity and memory of the antibody response. The serum of humans contains an efficient trypanolytic factor, the membrane pore-forming protein apolipoprotein L1 (APOL1). In the two human-infective trypanosomes, specific parasite resistance factors inhibit APOL1 activity. In turn, many African individuals express APOL1 variants that counteract these resistance factors, enabling them to avoid sleeping sickness. However, these variants are associated with chronic kidney disease, particularly in the context of virus-induced inflammation such as coronavirus disease 2019. Vaccination perspectives are discussed.
Collapse
Affiliation(s)
- Etienne Pays
- Laboratory of Molecular Parasitology, Université Libre de Bruxelles, Gosselies, Belgium;
| | - Magdalena Radwanska
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium;
| | - Stefan Magez
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea.,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; .,Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
4
|
Akoolo L, Rocha SC, Parveen N. Protozoan co-infections and parasite influence on the efficacy of vaccines against bacterial and viral pathogens. Front Microbiol 2022; 13:1020029. [PMID: 36504775 PMCID: PMC9732444 DOI: 10.3389/fmicb.2022.1020029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 11/07/2022] [Indexed: 11/26/2022] Open
Abstract
A wide range of protozoan pathogens either transmitted by vectors (Plasmodium, Babesia, Leishmania and Trypanosoma), by contaminated food or water (Entamoeba and Giardia), or by sexual contact (Trichomonas) invade various organs in the body and cause prominent human diseases, such as malaria, babesiosis, leishmaniasis, trypanosomiasis, diarrhea, and trichomoniasis. Humans are frequently exposed to multiple pathogens simultaneously, or sequentially in the high-incidence regions to result in co-infections. Consequently, synergistic or antagonistic pathogenic effects could occur between microbes that also influences overall host responses and severity of diseases. The co-infecting organisms can also follow independent trajectory. In either case, co-infections change host and pathogen metabolic microenvironments, compromise the host immune status, and affect microbial pathogenicity to influence tissue colonization. Immunomodulation by protozoa often adversely affects cellular and humoral immune responses against co-infecting bacterial pathogens and promotes bacterial persistence, and result in more severe disease symptoms. Although co-infections by protozoa and viruses also occur in humans, extensive studies are not yet conducted probably because of limited animal model systems available that can be used for both groups of pathogens. Immunosuppressive effects of protozoan infections can also attenuate vaccines efficacy, weaken immunological memory development, and thus attenuate protection against co-infecting pathogens. Due to increasing occurrence of parasitic infections, roles of acute to chronic protozoan infection on immunological changes need extensive investigations to improve understanding of the mechanistic details of specific immune responses alteration. In fact, this phenomenon should be seriously considered as one cause of breakthrough infections after vaccination against both bacterial and viral pathogens, and for the emergence of drug-resistant bacterial strains. Such studies would facilitate development and implementation of effective vaccination and treatment regimens to prevent or significantly reduce breakthrough infections.
Collapse
Affiliation(s)
- Lavoisier Akoolo
- Biorepository and Tissue Research Facility, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Sandra C. Rocha
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Nikhat Parveen
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, United States,*Correspondence: Nikhat Parveen,
| |
Collapse
|
5
|
Romero-Ramirez A, Casas-Sánchez A, Autheman D, Duffy CW, Brandt C, Clare S, Harcourt K, André MR, de Almeida Castilho Neto KJG, Teixeira MMG, Machado RZ, Coombes J, Flynn RJ, Wright GJ, Jackson AP. Vivaxin genes encode highly immunogenic, non-variant antigens on the Trypanosoma vivax cell-surface. PLoS Negl Trop Dis 2022; 16:e0010791. [PMID: 36129968 PMCID: PMC9529106 DOI: 10.1371/journal.pntd.0010791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 10/03/2022] [Accepted: 09/06/2022] [Indexed: 12/02/2022] Open
Abstract
Trypanosoma vivax is a unicellular hemoparasite, and a principal cause of animal African trypanosomiasis (AAT), a vector-borne and potentially fatal livestock disease across sub-Saharan Africa. Previously, we identified diverse T. vivax-specific genes that were predicted to encode cell surface proteins. Here, we examine the immune responses of naturally and experimentally infected hosts to these unique parasite antigens, to identify immunogens that could become vaccine candidates. Immunoprofiling of host serum shows that one particular family (Fam34) elicits a consistent IgG antibody response. This gene family, which we now call Vivaxin, encodes at least 124 transmembrane glycoproteins that display quite distinct expression profiles and patterns of genetic variation. We focused on one gene (viv-β8) that encodes one particularly immunogenic vivaxin protein and which is highly expressed during infections but displays minimal polymorphism across the parasite population. Vaccination of mice with VIVβ8 adjuvanted with Quil-A elicits a strong, balanced immune response and delays parasite proliferation in some animals but, ultimately, it does not prevent disease. Although VIVβ8 is localized across the cell body and flagellar membrane, live immunostaining indicates that VIVβ8 is largely inaccessible to antibody in vivo. However, our phylogenetic analysis shows that vivaxin includes other antigens shown recently to induce immunity against T. vivax. Thus, the introduction of vivaxin represents an important advance in our understanding of the T. vivax cell surface. Besides being a source of proven and promising vaccine antigens, the gene family is clearly an important component of the parasite glycocalyx, with potential to influence host-parasite interactions. Animal African trypanosomiasis (AAT) is an important livestock disease throughout sub-Saharan Africa and beyond. AAT is caused by Trypanosoma vivax, among other species, a unicellular parasite that is spread by biting tsetse flies and multiplies in the bloodstream and other tissues, leading to often fatal neurological conditions if untreated. Although concerted drug treatment and vector eradication programmes have succeeded in controlling Human African trypanosomiasis, AAT continues to adversely affect animal health and impede efficient food production and economic development in many less-developed countries. In this study, we attempted to identify parasite surface proteins that stimulated the strongest immune responses in naturally infected animals, as the basis for a vaccine. We describe the discovery of a new, species-specific protein family in T. vivax, which we call vivaxin. We show that one vivaxin protein (VIVβ8) is surface expressed and retards parasite proliferation when used to immunize mice, but does not prevent infection. Nevertheless, we also reveal that vivaxin includes another protein previously shown to induce protective immunity (IFX/VIVβ1). Besides its great potential for novel approaches to AAT control, the vivaxin family is revealed as a significant component of the T. vivax cell surface and may have important, species-specific roles in host interactions.
Collapse
Affiliation(s)
- Alessandra Romero-Ramirez
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Aitor Casas-Sánchez
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Delphine Autheman
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York, York, United Kingdom
| | - Craig W. Duffy
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Cordelia Brandt
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Simon Clare
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Katherine Harcourt
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Marcos Rogério André
- Department of Pathology, Reproduction and One Health, Faculty of Agrarian and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal, Sao Paulo, Brazil
| | - Kayo José Garcia de Almeida Castilho Neto
- Department of Pathology, Reproduction and One Health, Faculty of Agrarian and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal, Sao Paulo, Brazil
| | - Marta M. G. Teixeira
- Department of Parasitology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Sao Paulo, Brazil
| | - Rosangela Zacharias Machado
- Department of Pathology, Reproduction and One Health, Faculty of Agrarian and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal, Sao Paulo, Brazil
| | - Janine Coombes
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- School of Pharmacy and Life Sciences, The Robert Gordon University, Aberdeen, United Kingdom
| | - Robin J. Flynn
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Waterford Institute of Technology, Waterford, Ireland
| | - Gavin J. Wright
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York, York, United Kingdom
| | - Andrew P. Jackson
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- * E-mail:
| |
Collapse
|
6
|
Rajavel A, Klees S, Hui Y, Schmitt AO, Gültas M. Deciphering the Molecular Mechanism Underlying African Animal Trypanosomiasis by Means of the 1000 Bull Genomes Project Genomic Dataset. BIOLOGY 2022; 11:biology11050742. [PMID: 35625470 PMCID: PMC9138820 DOI: 10.3390/biology11050742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/05/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022]
Abstract
Simple Summary Climate change is increasing the risk of spreading vector-borne diseases such as African Animal Trypanosomiasis (AAT), which is causing major economic losses, especially in sub-Saharan African countries. Mainly considering this disease, we have investigated transcriptomic and genomic data from two cattle breeds, namely Boran and N‘Dama, where the former is known for its susceptibility and the latter one for its tolerance to the AAT. Despite the rich literature on this disease, there is still a need to investigate underlying genetic mechanisms to decipher the complex interplay of regulatory SNPs (rSNPs), their corresponding gene expression profiles and the downstream effectors associated with the AAT disease. The findings of this study complement our previous results, which mainly involve the upstream events, including transcription factors (TFs) and their co-operations as well as master regulators. Moreover, our investigation of significant rSNPs and effectors found in the liver, spleen and lymph node tissues of both cattle breeds could enhance the understanding of distinct mechanisms leading to either resistance or susceptibility of cattle breeds. Abstract African Animal Trypanosomiasis (AAT) is a neglected tropical disease and spreads by the vector tsetse fly, which carries the infectious Trypanosoma sp. in their saliva. Particularly, this parasitic disease affects the health of livestock, thereby imposing economic constraints on farmers, costing billions of dollars every year, especially in sub-Saharan African countries. Mainly considering the AAT disease as a multistage progression process, we previously performed upstream analysis to identify transcription factors (TFs), their co-operations, over-represented pathways and master regulators. However, downstream analysis, including effectors, corresponding gene expression profiles and their association with the regulatory SNPs (rSNPs), has not yet been established. Therefore, in this study, we aim to investigate the complex interplay of rSNPs, corresponding gene expression and downstream effectors with regard to the AAT disease progression based on two cattle breeds: trypanosusceptible Boran and trypanotolerant N’Dama. Our findings provide mechanistic insights into the effectors involved in the regulation of several signal transduction pathways, thereby differentiating the molecular mechanism with regard to the immune responses of the cattle breeds. The effectors and their associated genes (especially MAPKAPK5, CSK, DOK2, RAC1 and DNMT1) could be promising drug candidates as they orchestrate various downstream regulatory cascades in both cattle breeds.
Collapse
Affiliation(s)
- Abirami Rajavel
- Breeding Informatics Group, Department of Animal Sciences, Georg-August University, Margarethe von Wrangell-Weg 7, 37075 Göttingen, Germany; (S.K.); (Y.H.); (A.O.S.)
- Center for Integrated Breeding Research (CiBreed), Georg-August University, Carl-Sprengel-Weg 1, 37075 Göttingen, Germany
- Correspondence: (A.R.); (M.G.)
| | - Selina Klees
- Breeding Informatics Group, Department of Animal Sciences, Georg-August University, Margarethe von Wrangell-Weg 7, 37075 Göttingen, Germany; (S.K.); (Y.H.); (A.O.S.)
- Center for Integrated Breeding Research (CiBreed), Georg-August University, Carl-Sprengel-Weg 1, 37075 Göttingen, Germany
| | - Yuehan Hui
- Breeding Informatics Group, Department of Animal Sciences, Georg-August University, Margarethe von Wrangell-Weg 7, 37075 Göttingen, Germany; (S.K.); (Y.H.); (A.O.S.)
| | - Armin Otto Schmitt
- Breeding Informatics Group, Department of Animal Sciences, Georg-August University, Margarethe von Wrangell-Weg 7, 37075 Göttingen, Germany; (S.K.); (Y.H.); (A.O.S.)
- Center for Integrated Breeding Research (CiBreed), Georg-August University, Carl-Sprengel-Weg 1, 37075 Göttingen, Germany
| | - Mehmet Gültas
- Center for Integrated Breeding Research (CiBreed), Georg-August University, Carl-Sprengel-Weg 1, 37075 Göttingen, Germany
- Faculty of Agriculture, South Westphalia University of Applied Sciences, Lübecker Ring 2, 59494 Soest, Germany
- Correspondence: (A.R.); (M.G.)
| |
Collapse
|
7
|
|
8
|
Wait LF, Dobson AP, Graham AL. Do parasite infections interfere with immunisation? A review and meta-analysis. Vaccine 2020; 38:5582-5590. [DOI: 10.1016/j.vaccine.2020.06.064] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/12/2020] [Accepted: 06/21/2020] [Indexed: 12/18/2022]
|
9
|
Magez S, Pinto Torres JE, Obishakin E, Radwanska M. Infections With Extracellular Trypanosomes Require Control by Efficient Innate Immune Mechanisms and Can Result in the Destruction of the Mammalian Humoral Immune System. Front Immunol 2020; 11:382. [PMID: 32218784 PMCID: PMC7078162 DOI: 10.3389/fimmu.2020.00382] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/18/2020] [Indexed: 12/17/2022] Open
Abstract
Salivarian trypanosomes are extracellular parasites that affect humans, livestock, and game animals around the world. Through co-evolution with the mammalian immune system, trypanosomes have developed defense mechanisms that allow them to thrive in blood, lymphoid vessels, and tissue environments such as the brain, the fat tissue, and testes. Trypanosomes have developed ways to circumvent antibody-mediated killing and block the activation of the lytic arm of the complement pathway. Hence, this makes the innate immune control of the infection a crucial part of the host-parasite interaction, determining infection susceptibility, and parasitemia control. Indeed, trypanosomes use a combination of several independent mechanisms to avoid clearance by the humoral immune system. First, perpetuated antigenic variation of the surface coat allows to escape antibody-mediated elimination. Secondly, when antibodies bind to the coat, they are efficiently transported toward the endocytosis pathway, where they are removed from the coat proteins. Finally, trypanosomes engage in the active destruction of the mammalian humoral immune response. This provides them with a rescue solution in case antigenic variation does not confer total immunological invisibility. Both antigenic variation and B cell destruction pose significant hurdles for the development of anti-trypanosome vaccine strategies. However, developing total immune escape capacity and unlimited growth capabilities within a mammalian host is not beneficial for any parasite, as it will result in the accelerated death of the host itself. Hence, trypanosomes have acquired a system of quorum sensing that results in density-dependent population growth arrest in order to prevent overpopulating the host. The same system could possibly sense the infection-associated host tissue damage resulting from inflammatory innate immune responses, in which case the quorum sensing serves to prevent excessive immunopathology and as such also promotes host survival. In order to put these concepts together, this review summarizes current knowledge on the interaction between trypanosomes and the mammalian innate immune system, the mechanisms involved in population growth regulation, antigenic variation and the immuno-destructive effect of trypanosomes on the humoral immune system. Vaccine trials and a discussion on the role of innate immune modulation in these trials are discussed at the end.
Collapse
Affiliation(s)
- Stefan Magez
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea.,Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium.,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Emmanuel Obishakin
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea.,Biotechnology Division, National Veterinary Research Institute, Vom, Nigeria
| | - Magdalena Radwanska
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
10
|
Silva Pereira S, de Almeida Castilho Neto KJG, Duffy CW, Richards P, Noyes H, Ogugo M, Rogério André M, Bengaly Z, Kemp S, Teixeira MMG, Machado RZ, Jackson AP. Variant antigen diversity in Trypanosoma vivax is not driven by recombination. Nat Commun 2020; 11:844. [PMID: 32051413 PMCID: PMC7015903 DOI: 10.1038/s41467-020-14575-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 01/18/2020] [Indexed: 11/09/2022] Open
Abstract
African trypanosomes (Trypanosoma) are vector-borne haemoparasites that survive in the vertebrate bloodstream through antigenic variation of their Variant Surface Glycoprotein (VSG). Recombination, or rather segmented gene conversion, is fundamental in Trypanosoma brucei for both VSG gene switching and for generating antigenic diversity during infections. Trypanosoma vivax is a related, livestock pathogen whose VSG lack structures that facilitate gene conversion in T. brucei and mechanisms underlying its antigenic diversity are poorly understood. Here we show that species-wide VSG repertoire is broadly conserved across diverse T. vivax clinical strains and has limited antigenic repertoire. We use variant antigen profiling, coalescent approaches and experimental infections to show that recombination plays little role in diversifying T. vivax VSG sequences. These results have immediate consequences for both the current mechanistic model of antigenic variation in African trypanosomes and species differences in virulence and transmission, requiring reconsideration of the wider epidemiology of animal African trypanosomiasis.
Collapse
Affiliation(s)
- Sara Silva Pereira
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, 146 Brownlow Hill, Liverpool, L3 5RF, UK
| | - Kayo J G de Almeida Castilho Neto
- Department of Veterinary Pathology, Faculty of Agrarian and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal, SP, Brazil
| | - Craig W Duffy
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, 146 Brownlow Hill, Liverpool, L3 5RF, UK
| | - Peter Richards
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, 146 Brownlow Hill, Liverpool, L3 5RF, UK
| | - Harry Noyes
- Institute of Integrative Biology, University of Liverpool, Biosciences Building, Crown Street, Liverpool, L69 7ZB, UK
| | - Moses Ogugo
- Livestock Genetic Programme, International Livestock Research Institute, 30709 Naivasha Road, Nairobi, Kenya
| | - Marcos Rogério André
- Department of Veterinary Pathology, Faculty of Agrarian and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal, SP, Brazil
| | - Zakaria Bengaly
- International Research Centre for Livestock Development in the Sub-humid Zone (CIRDES), No. 559, rue 5-31 angle, Avenue du Gouverneur Louveau, Bobo-Dioulasso, Burkina Faso
| | - Steve Kemp
- Livestock Genetic Programme, International Livestock Research Institute, 30709 Naivasha Road, Nairobi, Kenya
| | - Marta M G Teixeira
- Department of Parasitology, Institute of Biomedical Sciences, University of Sao Paulo, Avenue Professor Lineu Prestes, 1374 Cidade Universitaria, Sao Paulo, SP, 05508-000, Brazil
| | - Rosangela Z Machado
- Department of Veterinary Pathology, Faculty of Agrarian and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal, SP, Brazil
| | - Andrew P Jackson
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, 146 Brownlow Hill, Liverpool, L3 5RF, UK.
| |
Collapse
|
11
|
Radwanska M, Vereecke N, Deleeuw V, Pinto J, Magez S. Salivarian Trypanosomosis: A Review of Parasites Involved, Their Global Distribution and Their Interaction With the Innate and Adaptive Mammalian Host Immune System. Front Immunol 2018; 9:2253. [PMID: 30333827 PMCID: PMC6175991 DOI: 10.3389/fimmu.2018.02253] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/11/2018] [Indexed: 01/27/2023] Open
Abstract
Salivarian trypanosomes are single cell extracellular parasites that cause infections in a wide range of hosts. Most pathogenic infections worldwide are caused by one of four major species of trypanosomes including (i) Trypanosoma brucei and the human infective subspecies T. b. gambiense and T. b. rhodesiense, (ii) Trypanosoma evansi and T. equiperdum, (iii) Trypanosoma congolense and (iv) Trypanosoma vivax. Infections with these parasites are marked by excessive immune dysfunction and immunopathology, both related to prolonged inflammatory host immune responses. Here we review the classification and global distribution of these parasites, highlight the adaptation of human infective trypanosomes that allow them to survive innate defense molecules unique to man, gorilla, and baboon serum and refer to the discovery of sexual reproduction of trypanosomes in the tsetse vector. With respect to the immunology of mammalian host-parasite interactions, the review highlights recent findings with respect to the B cell destruction capacity of trypanosomes and the role of T cells in the governance of infection control. Understanding infection-associated dysfunction and regulation of both these immune compartments is crucial to explain the continued failures of anti-trypanosome vaccine developments as well as the lack of any field-applicable vaccine based anti-trypanosomosis intervention strategy. Finally, the link between infection-associated inflammation and trypanosomosis induced anemia is covered in the context of both livestock and human infections.
Collapse
Affiliation(s)
- Magdalena Radwanska
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea
| | - Nick Vereecke
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea.,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Violette Deleeuw
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Joar Pinto
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Stefan Magez
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea.,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
12
|
Onyilagha C, Jia P, Jayachandran N, Hou S, Okwor I, Kuriakose S, Marshall A, Uzonna JE. The B cell adaptor molecule Bam32 is critically important for optimal antibody response and resistance to Trypanosoma congolense infection in mice. PLoS Negl Trop Dis 2015; 9:e0003716. [PMID: 25875604 PMCID: PMC4395458 DOI: 10.1371/journal.pntd.0003716] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 03/21/2015] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Bam32, a 32 kDa adaptor molecule, plays important role in B cell receptor signalling, T cell receptor signalling and antibody affinity maturation in germinal centres. Since antibodies against trypanosome variant surface glycoproteins (VSG) are critically important for control of parasitemia, we hypothesized that Bam32 deficient (Bam32-/-) mice would be susceptible to T. congolense infection. METHODOLOGY/PRINCIPAL FINDINGS We found that T. congolense-infected Bam32-/- mice successfully control the first wave of parasitemia but then fail to control subsequent waves and ultimately succumb to their infection unlike wild type (WT) C57BL6 mice which are relatively resistant. Although infected Bam32-/- mice had significantly higher hepatomegaly and splenomegaly, their serum AST and ALT levels were not different, suggesting that increased liver pathology may not be responsible for the increased susceptibility of Bam32-/- mice to T. congolense. Using direct ex vivo flow cytometry and ELISA, we show that CD4+ T cells from infected Bam32-/- mice produced significantly increased amounts of disease-exacerbating proinflammatory cytokines (including IFN-γ, TNF-α and IL-6). However, the percentages of regulatory T cells and IL-10-producing CD4+ cells were similar in infected WT and Bam32-/- mice. While serum levels of parasite-specific IgM antibodies were normal, the levels of parasite-specific IgG, (particularly IgG1 and IgG2a) were significantly lower in Bam32-/- mice throughout infection. This was associated with impaired germinal centre response in Bam32-/- mice despite increased numbers of T follicular helper (Tfh) cells. Adoptive transfer studies indicate that intrinsic B cell defect was responsible for the enhanced susceptibility of Bam32-/- mice to T. congolense infection. CONCLUSIONS/SIGNIFICANCE Collectively, our data show that Bam32 is important for optimal anti-trypanosome IgG antibody response and suppression of disease-promoting proinflammatory cytokines and its deficiency leads to inability to control T. congolense infection in mice.
Collapse
Affiliation(s)
- Chukwunonso Onyilagha
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ping Jia
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Nipun Jayachandran
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sen Hou
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ifeoma Okwor
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Shiby Kuriakose
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Aaron Marshall
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jude E. Uzonna
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- * E-mail:
| |
Collapse
|
13
|
Lejon V, Mumba Ngoyi D, Kestens L, Boel L, Barbé B, Kande Betu V, van Griensven J, Bottieau E, Muyembe Tamfum JJ, Jacobs J, Büscher P. Gambiense human african trypanosomiasis and immunological memory: effect on phenotypic lymphocyte profiles and humoral immunity. PLoS Pathog 2014; 10:e1003947. [PMID: 24603894 PMCID: PMC3946376 DOI: 10.1371/journal.ppat.1003947] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 01/09/2014] [Indexed: 12/25/2022] Open
Abstract
In mice, experimental infection with Trypanosoma brucei causes decreased bone marrow B-cell development, abolished splenic B-cell maturation and loss of antibody mediated protection including vaccine induced memory responses. Nothing is known about this phenomenon in human African trypanosomiasis (HAT), but if occurring, it would imply the need of revaccination of HAT patients after therapy and abolish hope for a HAT vaccine. The effect of gambiense HAT on peripheral blood memory T- and B-cells and on innate and vaccine induced antibody levels was examined. The percentage of memory B- and T-cells was quantified in peripheral blood, prospectively collected in DR Congo from 117 Trypanosoma brucei gambiense infected HAT patients before and six months after treatment and 117 controls at the same time points. Antibodies against carbohydrate antigens on red blood cells and against measles were quantified. Before treatment, significantly higher percentages of memory B-cells, mainly T-independent memory B-cells, were observed in HAT patients compared to controls (CD20+CD27+IgM+, 13.0% versus 2.0%, p<0.001). The percentage of memory T-cells, mainly early effector/memory T-cells, was higher in HAT (CD3+CD45RO+CD27+, 19.4% versus 16.7%, p = 0.003). After treatment, the percentage of memory T-cells normalized, the percentage of memory B-cells did not. The median anti-red blood cell carbohydrate IgM level was one titer lower in HAT patients than in controls (p<0.004), and partially normalized after treatment. Anti-measles antibody concentrations were lower in HAT patients than in controls (medians of 1500 versus 2250 mIU/ml, p = 0.02), and remained so after treatment, but were above the cut-off level assumed to provide protection in 94.8% of HAT patients, before and after treatment (versus 98.3% of controls, p = 0.3). Although functionality of the B-cells was not verified, the results suggest that immunity was conserved in T.b. gambiense infected HAT patients and that B-cell dysfunction might not be that severe as in mouse models.
Collapse
Affiliation(s)
- Veerle Lejon
- Institute of Tropical Medicine, Antwerp, Belgium
- Institut de Recherche pour le Développement, UMR 177 IRD-CIRAD INTERTRYP, Montpellier, France
| | - Dieudonné Mumba Ngoyi
- Institut National de Recherche Biomédicale, Kinshasa, Democratic Republic of the Congo
| | - Luc Kestens
- Institute of Tropical Medicine, Antwerp, Belgium
| | - Luc Boel
- Institute of Tropical Medicine, Antwerp, Belgium
| | | | - Victor Kande Betu
- Programme Nationale de Lutte contre la Trypanosomiase Humaine Africaine, Kinshasa, Democratic Republic of the Congo
| | | | | | | | - Jan Jacobs
- Institute of Tropical Medicine, Antwerp, Belgium
| | | |
Collapse
|
14
|
Ng DHL, Skehel JJ, Kassiotis G, Langhorne J. Recovery of an antiviral antibody response following attrition caused by unrelated infection. PLoS Pathog 2014; 10:e1003843. [PMID: 24391499 PMCID: PMC3879355 DOI: 10.1371/journal.ppat.1003843] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 11/05/2013] [Indexed: 01/28/2023] Open
Abstract
The homeostatic mechanisms that regulate the maintenance of immunological memory to the multiple pathogen encounters over time are unknown. We found that a single malaria episode caused significant dysregulation of pre-established Influenza A virus-specific long-lived plasma cells (LLPCs) resulting in the loss of Influenza A virus-specific Abs and increased susceptibility to Influenza A virus re-infection. This loss of LLPCs involved an FcγRIIB-dependent mechanism, leading to their apoptosis. However, given enough time following malaria, the LLPC pool and humoral immunity to Influenza A virus were eventually restored. Supporting a role for continuous conversion of Influenza A virus-specific B into LLPCs in the restoration of Influenza A virus immunity, B cell depletion experiments also demonstrated a similar requirement for the long-term maintenance of serum Influenza A virus-specific Abs in an intact LLPC compartment. These findings show that, in addition to their established role in the anamnestic response to reinfection, the B cell pool continues to be a major contributor to the maintenance of long-term humoral immunity following primary Influenza A virus infection, and to the recovery from attrition following heterologous infection. These data have implications for understanding the longevity of protective efficacy of vaccinations in countries where continuous infections are endemic. Antibody responses to infectious pathogens are critical in host survival, recovery and protection from reinfection; they also correlate with the success of vaccination. It is currently thought that antibody serum titers are maintained at protective levels over long periods of time by specialized long-lived antibody-secreting plasma cells residing in the bone marrow. Indeed, antibodies against the original virus can still be found in survivors of the 1918 Spanish Flu, more than 90 years ago. However, it is also becoming clear that subsequent infection with heterologous pathogens may cause attrition of previously established immunological memory, in order to accommodate new lymphocyte specificities in the finite space of the host. This phenomenon is seemingly at odds with long-term maintenance of immunological memory. We also show that a single episode of malaria, caused by infection by Plasmodium chabaudi, leads to the loss of preexisting plasma cells, serum antibodies and protective immunity against Influenza A virus. However, Influenza A virus-specific immunity does eventually recover in these animals with the replenishment of plasma cells by B cells over the course of several weeks. Thus, the reported mechanism reconciles attrition of immunological memory by heterologous infection and long-term stability, and places B cells, instead of their descendant plasma cells, at the center of humoral memory.
Collapse
Affiliation(s)
- Dorothy H. L. Ng
- Division of Immunoregulation, MRC National Institute for Medical Research, London, United Kingdom
- Division of Parasitology, MRC National Institute for Medical Research, London, United Kingdom
| | - John J. Skehel
- Division of Virology, MRC National Institute for Medical Research, London, United Kingdom
| | - George Kassiotis
- Division of Immunoregulation, MRC National Institute for Medical Research, London, United Kingdom
- * E-mail: (GK); (JL)
| | - Jean Langhorne
- Division of Parasitology, MRC National Institute for Medical Research, London, United Kingdom
- * E-mail: (GK); (JL)
| |
Collapse
|
15
|
Widespread trypanosome infections in a population of eastern hellbenders (Cryptobranchus alleganiensis alleganiensis) in Virginia, USA. Parasitol Res 2012; 112:453-6. [PMID: 22926647 DOI: 10.1007/s00436-012-3076-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 08/06/2012] [Indexed: 10/28/2022]
|
16
|
Okwor I, Onyilagha C, Kuriakose S, Mou Z, Jia P, Uzonna JE. Regulatory T cells enhance susceptibility to experimental Trypanosoma congolense infection independent of mouse genetic background. PLoS Negl Trop Dis 2012; 6:e1761. [PMID: 22860150 PMCID: PMC3409116 DOI: 10.1371/journal.pntd.0001761] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 06/20/2012] [Indexed: 12/22/2022] Open
Abstract
Background BALB/c mice are highly susceptible while C57BL/6 are relatively resistant to experimental Trypanosoma congolense infection. Although regulatory T cells (Tregs) have been shown to regulate the pathogenesis of experimental T. congolense infection, their exact role remains controversial. We wished to determine whether Tregs contribute to distinct phenotypic outcomes in BALB/c and C57BL/6 mice and if so how they operate with respect to control of parasitemia and production of disease-exacerbating proinflammatory cytokines. Methodology/Findings BALB/c and C57BL/6 mice were infected intraperitoneally (i.p) with 103T. congolense clone TC13 and both the kinetics of Tregs expansion and intracellular cytokine profiles in the spleens and livers were monitored directly ex vivo by flow cytometry. In some experiments, mice were injected with anti-CD25 mAb prior or post T. congolense infection or adoptively (by intravenous route) given highly enriched naïve CD25+ T lymphocytes prior to T. congolense infection and the inflammatory cytokine/chemokine levels and survival were monitored. In contrast to a transient and non significant increase in the percentages and absolute numbers of CD4+CD25+Foxp3+ T cells (Tregs) in C57BL/6 mouse spleens and livers, a significant increase in the percentage and absolute numbers of Tregs was observed in spleens of infected BALB/c mice. Ablation or increasing the number of CD25+ cells in the relatively resistant C57BL/6 mice by anti-CD25 mAb treatment or by adoptive transfer of CD25+ T cells, respectively, ameliorates or exacerbates parasitemia and production of proinflammatory cytokines. Conclusion Collectively, our results show that regulatory T cells contribute to susceptibility in experimental murine trypanosomiasis in both the highly susceptible BALB/c and relatively resistant C57BL/6 mice. BALB/c mice are highly susceptible while C57BL/6 is relatively resistant to experimental Trypanosoma congolense infection. Acute death observed in infected BALB/c mice is usually associated with the excessive production of pro-inflammatory cytokines. Regulatory T cells (Tregs) have been shown to play a significant role in the pathogenesis of many diseases including those caused by parasites. However, the role of Tregs in the pathogenesis of T. congolense infection remains unclear. We were interested in addressing the following questions: Do Tregs contribute to the distinct phenotypic outcomes observed in T. congolense-infected BALB/c and C57BL/6 mice? If so, where and how do they operate with respect to parasitemia and cytokine response? By selectively altering the numbers of these cells either by targeted depletion with monoclonal antibody or adoptive transfer of highly enriched naïve CD25+ cells prior to infection, we show that Tregs impairs efficient parasite control and impacts on production of disease-exacerbating proinflammatory cytokines. Collectively, our findings suggest that Tregs contribute to enhanced susceptibility to experimental T. congolense infection in mice.
Collapse
Affiliation(s)
- Ifeoma Okwor
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Chukwunonso Onyilagha
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Shiby Kuriakose
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Zhirong Mou
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ping Jia
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jude E. Uzonna
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- * E-mail:
| |
Collapse
|
17
|
The potential impact of native Australian trypanosome infections on the health of koalas (Phascolarctos cinereus). Parasitology 2011; 138:873-83. [PMID: 21524321 DOI: 10.1017/s0031182011000369] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Whole blood collected from koalas admitted to the Australian Zoo Wildlife Hospital (AZWH), Beerwah, QLd, Australia, during late 2006-2009 was tested using trypanosome species-specific 18S rDNA PCRs designed to amplify DNA from Trypanosoma irwini, T. gilletti and T. copemani. Clinical records for each koala sampled were reviewed and age, sex, blood packed cell volume (PCV), body condition, signs of illness, blood loss, trauma, chlamydiosis, bone marrow disease, koala AIDS and hospital admission outcome ('survival'/ 'non-survival') were correlated with PCR results. Overall 73.8% (439/595) of the koalas were infected with at least 1 species of trypanosome. Trypanosoma irwini was detected in 423/595 (71.1%), T. gilletti in 128/595 (21.5%) and T. copemani in 26/595 (4.4%) of koalas. Mixed infections were detected in 125/595 (21%) with co-infections of T. irwini and T. gilletti (101/595, 17%) being most common. There was a statistical association between infection with T. gilletti with lower PCV values and body condition scores in koalas with signs of chlamydiosis, bone marrow disease or koala AIDS. No association between T. gilletti infection and any indicator of health was observed in koalas without signs of concurrent disease. This raises the possibility that T. gilletti may be potentiating other disease syndromes affecting koalas.
Collapse
|
18
|
Abstract
SUMMARYAnti-trypanosomiasis vaccination still remains the best theoretical option in the fight against a disease that is continuously hovering between its wildlife reservoir and its reservoir in man and livestock. While antigentic variation of the parasite surface coat has been considered the major obstacle in the development of a functional vaccine, recent research into the biology of B cells has indicated that the problems might go further than that. This paper reviews past and current attempts to design both anti-trypanosome vaccines, as well as vaccines directed towards the inhibition of infection-associated pathology.
Collapse
|
19
|
Magez S, Radwanska M. African trypanosomiasis and antibodies: implications for vaccination, therapy and diagnosis. Future Microbiol 2010; 4:1075-87. [PMID: 19824795 DOI: 10.2217/fmb.09.65] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
African trypanosomiasis causes devastating effects on human populations and livestock herds in large parts of sub-Saharan Africa. Control of the disease is hampered by the lack of any efficient vaccination results in a field setting, and the severe side effects of current drug therapies. In addition, with the exception of Trypanosoma brucei gambiense infections, the diagnosis of trypanosomiasis has to rely on microscopic analysis of blood samples, as other specific tools are nonexistent. However, new developments in biotechnology, which include loop-mediated isothermal amplification as an adaptation to conventional PCR, as well as the antibody engineering that has allowed the development of Nanobody technology, offer new perspectives in both the detection and treatment of trypanosomiasis. In addition, recent data on parasite-induced B-cell memory destruction offer new insights into mechanisms of vaccine failure, and should lead us towards new strategies to overcome trypanosome defenses operating against the host immune system.
Collapse
Affiliation(s)
- Stefan Magez
- Department of Molecular & Cellular Interactions, Flanders Institute for Biotechnology, Rijvisschestraat 120, B-9052 Ghent, Belgium.
| | | |
Collapse
|
20
|
Host-parasite interactions in trypanosomiasis: on the way to an antidisease strategy. Infect Immun 2009; 77:1276-84. [PMID: 19168735 DOI: 10.1128/iai.01185-08] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
21
|
Radwanska M, Guirnalda P, De Trez C, Ryffel B, Black S, Magez S. Trypanosomiasis-induced B cell apoptosis results in loss of protective anti-parasite antibody responses and abolishment of vaccine-induced memory responses. PLoS Pathog 2008; 4:e1000078. [PMID: 18516300 PMCID: PMC2386555 DOI: 10.1371/journal.ppat.1000078] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2007] [Accepted: 04/25/2008] [Indexed: 02/02/2023] Open
Abstract
African trypanosomes of the Trypanosoma brucei species are extra-cellular parasites that cause human African trypanosomiasis (HAT) as well as infections in game animals and livestock. Trypanosomes are known to evade the immune response of their mammalian host by continuous antigenic variation of their surface coat. Here, we aim to demonstrate that in addition, trypanosomes (i) cause the loss of various B cell populations, (ii) disable the hosts' capacity to raise a long-lasting specific protective anti-parasite antibody response, and (iii) abrogate vaccine-induced protective response to a non-related human pathogen such as Bordetella pertussis. Using a mouse model for T. brucei, various B cell populations were analyzed by FACS at different time points of infection. The results show that during early onset of a T. brucei infection, spleen remodeling results in the rapid loss of the IgM+ marginal zone (IgM+MZ) B cell population characterized as B220+IgMHighIgDInt CD21HighCD23LowCD1d+CD138−. These cells, when isolated during the first peak of infection, stained positive for Annexin V and had increased caspase-3 enzyme activity. Elevated caspase-3 mRNA levels coincided with decreased mRNA levels of the anti-apoptotic Bcl-2 protein and BAFF receptor (BAFF-R), indicating the onset of apoptosis. Moreover, affected B cells became unresponsive to stimulation by BCR cross-linking with anti-IgM Fab fragments. In vivo, infection-induced loss of IgM+ B cells coincided with the disappearance of protective variant-specific T-independent IgM responses, rendering the host rapidly susceptible to re-challenge with previously encountered parasites. Finally, using the well-established human diphtheria, tetanus, and B. pertussis (DTPa) vaccination model in mice, we show that T. brucei infections abrogate vaccine-induced protective responses to a non-related pathogen such as B. pertussis. Infections with T. brucei parasites result in the rapid loss of T–cell independent IgM+MZ B cells that are normally functioning as the primary immune barrier against blood-borne pathogens. In addition, ongoing trypanosome infections results in the rapid loss of B cell responsiveness and prevent the induction of protective memory responses. Finally, trypanosome infections disable the host's capacity to recall vaccine-induced memory responses against non-related pathogens. In particular, these last results call for detailed studies of the effect of HAT on memory recall responses in humans, prior to the planning of any mass vaccination campaign in HAT endemic areas. African trypanosomes are extracellular parasites that cause the deadly disease sleeping sickness in humans, and nagana in cattle. The control of infection is believed to be largely dependent on the host antibody response. We postulate here that protective anti-trypanosome responses mainly involve splenic marginal zone B cells, as they are implicated in the production of antibodies against blood-borne pathogens. In this work, we show that trypanosome infections induce the rapid loss of these marginal zone B cells, coinciding with the loss of the splenic marginal zone itself. While the infection does result in the induction of plasma cell differentiation and antibody secretion, the loss of the marginal zone B cell population results in the loss of specific protective responses. In addition, we also show that host memory responses are destroyed during infection, even affecting unrelated vaccine-induced memory responses such as those induced by the commercially available DTPa vaccine. The latter finding is crucial for the evaluation of mass vaccination approaches in African regions where trypanosome infections are prevalent.
Collapse
Affiliation(s)
- Magdalena Radwanska
- Laboratoire de Parasitologie, Université Libre de Bruxelles, ULB, Brussels, Belgium
| | - Patrick Guirnalda
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts, United States of America
| | - Carl De Trez
- Laboratoire de Parasitologie, Université Libre de Bruxelles, ULB, Brussels, Belgium
| | - Bernard Ryffel
- Molecular Immunology and Embriology IEM2815, Centre National de la Recherche Scientifique, Orléans, France
| | - Samuel Black
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts, United States of America
| | - Stefan Magez
- Laboratory for Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Molecular and Cellular Interactions, VIB, Brussels, Belgium
- * E-mail:
| |
Collapse
|
22
|
Vincendeau P, Bouteille B. Immunology and immunopathology of African trypanosomiasis. AN ACAD BRAS CIENC 2006; 78:645-65. [PMID: 17143404 DOI: 10.1590/s0001-37652006000400004] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2005] [Accepted: 10/05/2005] [Indexed: 11/21/2022] Open
Abstract
Major modifications of immune system have been observed in African trypanosomiasis. These immune reactions do not lead to protection and are also involved in immunopathology disorders. The major surface component (variable surface glycoprotein,VSG) is associated with escape to immune reactions, cytokine network dysfunctions and autoantibody production. Most of our knowledge result from experimental trypanosomiasis. Innate resistance elements have been characterised. In infected mice, VSG preferentially stimulates a Th 1-cell subset. A response of <FONT FACE=Symbol>gd</FONT> and CD8 T cells to trypanosome antigens was observed in trypanotolerant cattle. An increase in CD5 B cells, responsible for most serum IgM and production of autoantibodies has been noted in infected cattle. Macrophages play important roles in trypanosomiasis, in synergy with antibodies (phagocytosis) and by secreting various molecules (radicals, cytokines, prostaglandins,...). Trypanosomes are highly sensitive to TNF-alpha, reactive oxygen and nitrogen intermediates. TNF-alpha is also involved in cachexia. IFN-gamma acts as a parasite growth factor. These various elements contribute to immunosuppression. Trypanosomes have learnt to use immune mechanisms to its own profit. Recent data show the importance of alternative macrophage activation, including arginase induction. L-ornithine produced by host arginase is essential to parasite growth. All these data reflect the deep insight into the immune system realised by trypanosomes and might suggest interference therapeutic approaches.
Collapse
|
23
|
Ihedioha JI, Chineme CN, Okoye JOA. The Leucocytic and Parasitaemic Profiles and Immune Response of Rats Treated with Retinyl Palmitate before Infection with Trypanosoma brucei. J Comp Pathol 2003; 129:241-50. [PMID: 14554122 DOI: 10.1016/s0021-9975(03)00039-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The effects of oral administration of retinyl palmitate (30,000 IU/kg bodyweight) for 3 days to rats before infection with Trypanosoma brucei were investigated by examining the leucocytic and parasitaemic profiles, and the antibody response to sheep red blood cells. The pretreatment significantly (P<0.01) improved the leucocytic profile (especially the absolute lymphocyte count) from day 7 post-infection to the time of death. It also significantly delayed the onset of parasitaemia (i.e., lengthened the pre-patent period) and led to reduced levels of parasitaemia throughout the period of infection. Pretreatment significantly increased the antibody response to sheep red blood cells (P<0.01) throughout the infection, even after treatment with diminazene aceturate, to levels that more than compensated for the immunosuppressive effect of the T. brucei. Further studies are warranted on the possible use of retinyl palmitate in overcoming immunosuppression associated with trypanosome infections in man and animals in endemic areas.
Collapse
Affiliation(s)
- J I Ihedioha
- Department of Veterinary Pathology and Microbiology, University of Nigeria, Nsukka, P O Box 3236, Nsukka, Enugu State, Nigeria
| | | | | |
Collapse
|
24
|
Holland WG, Do TT, Huong NT, Dung NT, Thanh NG, Vercruysse J, Goddeeris BM. The effect of Trypanosoma evansi infection on pig performance and vaccination against classical swine fever. Vet Parasitol 2003; 111:115-23. [PMID: 12531288 DOI: 10.1016/s0304-4017(02)00363-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Although Trypanosoma evansi is not considered as an important pathogen in pigs, it may interfere with other pathogens or vaccinations by its immunosuppressive nature. In order to determine whether T. evansi alters pig performance and induces immunosuppression in pigs, induction of immune responses by vaccination against classical swine fever (CSF) and by immunization with a control antigen, human serum albumin (HSA), was assessed in T. evansi-infected and non-infected animals. Although T. evansi infection did not have a significant influence on growth performance, feed conversion or PCV, antibody responses against both the test antigen HSA and the CSF vaccine were significantly reduced in T. evansi-infected animals as compared to uninfected animals. Moreover, the reduced response against the CSF vaccine appears to be accompanied by a less well-developed protection against CSF with higher fever responses and leukopenia. This immunosuppression might explain the accounts of poor protection of CSF-vaccinated pigs reported in T. evansi-endemic areas of Vietnam, and suggests that prior treatments with trypanocidal drugs to improve the efficacy of CSF vaccination, may be justified.
Collapse
Affiliation(s)
- W G Holland
- Ghent University, Faculty of Veterinary Medicine, Department Virology-Parasitology-Immunology, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | | | | | | | | | | | | |
Collapse
|
25
|
Chiejina SN, Wakelin D, Goyal PK. Trypanosome-induced modulation of responses to concurrent helminth infection. Res Vet Sci 2003; 74:47-53. [PMID: 12507566 DOI: 10.1016/s0034-5288(02)00152-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Infections with African trypanosomes are known to suppress immune responses to vaccines and to gastrointestinal nematode infections in livestock. Experimental infections with Trypanosoma brucei (Tb) and the gastrointestinal nematode Nippostrongylus brasiliensis (Nb) in mice were used to identify possible mechanisms involved in interference with anti-worm responses and to examine the effects of host genotype on the extent of suppression seen. Concurrent infections with T. brucei resulted in a prolongation of worm survival and a dramatic increase in faecal egg output. Infection also resulted in a marked suppression of the proliferative response of mesenteric lymphocytes (MLNC) to in vitro mitogenic stimulation. When MLNC from concurrently infected mice were stimulated in vitro with the mitogen ConA they released more IFN-gamma and less IL-5 than cells from mice infected only with N. brasiliensis. These data are interpreted in terms of a trypanosome-mediated influence on the development of host-protective type-2 T helper cell responses against N. brasiliensis. The degree to which T. brucei altered the kinetics of the nematode infection was influenced by the particular mouse strain concerned.
Collapse
Affiliation(s)
- S N Chiejina
- School of Life and Environmental Sciences, University of Nottingham, University Park, NG7 2RD, Nottingham, UK
| | | | | |
Collapse
|
26
|
Holland WG, My LN, Dung TV, Thanh NG, Tam PT, Vercruysse J, Goddeeris BM. The influence of T. evansi infection on the immuno-responsiveness of experimentally infected water buffaloes. Vet Parasitol 2001; 102:225-34. [PMID: 11777602 DOI: 10.1016/s0304-4017(01)00534-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
In order to define the immuno-suppressive capacity of Trypanosoma evansi infections in buffaloes on the induction of immune responses against heterologous antigens, infected and non-infected buffaloes were vaccinated against Pasteurella multocida (haemorrhagic septicemia) and were simultaneously immunised with a control antigen, human serum albumin (HSA). Antibody responses against HSA were significantly reduced in T. evansi-infected animals, but no conclusive data were obtained on the antibody responses against P. multocida. Conversely, the local inflammatory response at the site of Pasteurella vaccination, as measured by increase in size, was significantly reduced in T. evansi-infected animals. These results indicate that the inductive capacity to mount humoral and cell-mediated immune responses against heterologous antigens is suppressed in T. evansi-infected animals. Consequently, T. evansi infection might interfere with the development of protective immunity upon heterologous vaccinations and could explain the poor protection of Pasteurella-vaccinated buffaloes in T. evansi-endemic areas of Vietnam.
Collapse
Affiliation(s)
- W G Holland
- Faculty of Veterinary Medicine, Laboratory of Parasitology, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium
| | | | | | | | | | | | | |
Collapse
|
27
|
Tabel H, Kaushik RS, Uzonna JE. Susceptibility and resistance to Trypanosoma congolense infections. Microbes Infect 2000; 2:1619-29. [PMID: 11113381 DOI: 10.1016/s1286-4579(00)01318-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have put emphasis on recent findings in experimental Trypanosoma congolense infections in highly susceptible BALB/c and relatively resistant C57Bl/6 mice. Based on various analyses, it has been shown that a major difference in resistance to T. congolense infections is expressed early in infection at the macrophage level. A novel plastic-adherent Thy1.2(+) suppressor lymphocyte, which in absolute synergy with a Thy 1.2(-) cell exerts its suppression via interleukin-10 and interferon-gamma opens up an exciting new field of research.
Collapse
Affiliation(s)
- H Tabel
- Department of Veterinary Microbiology, University of Saskatchewan, SK, S7N 5B4, Saskatoon, Canada.
| | | | | |
Collapse
|
28
|
Mattioli RC, Pandey VS, Murray M, Fitzpatrick JL. Immunogenetic influences on tick resistance in African cattle with particular reference to trypanotolerant N'Dama (Bos taurus) and trypanosusceptible Gobra zebu (Bos indicus) cattle. Acta Trop 2000; 75:263-77. [PMID: 10838210 DOI: 10.1016/s0001-706x(00)00063-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In sub-Saharan Africa, tick infestation and tick-borne infections together with tsetse-transmitted trypanosomosis arguably constitute the main parasitological disease complex constraining livestock production. Resistance to tick attack and tick-borne micro-organisms (TBMs) varies among different breeds of cattle. The magnitude of losses due to these parasites is related to an extent to the degree of breed resistance. Generally, zebu (Bos indicus) cattle possess a higher resistance to ticks and TBMs than European (Bos taurus) cattle. The host's immune system would appear to be the single most important factor that regulates this resistance. This paper reports on the main effector immune mechanisms governing resistance against ticks and TBMs. The cellular immune response appears more effective and stable than humoral immunity in modulating resistance to ticks and TBMs. Similarities between the immune mechanisms employed by trypanotolerant N'Dama (B. taurus) cattle, when infected with trypanosomes, and those elicited by tick bites and TBMs seem to exist, particularly at the skin level in the early phases of parasitic invasion. Moreover, there is evidence that in the N'Dama breed, resistance against ticks per se also has a genetic basis. Therefore, the N'Dama appears to be a unique breed in that it exhibits resistance to several parasitic diseases and/or infections, including helminths, when compared to other cattle breeds in West Africa. It is concluded that the multi-parasite resistant traits of the N'Dama breed should be exploited in those areas where trypanosomosis, ticks and tick-borne diseases constrain animal production. This should be of benefit for low-input farming systems where the use of chemicals for prophylaxis and therapy is limited by their relatively high cost. Additionally, the potential contribution of multiple disease resistant N'Dama cattle should be considered in crossbreeding programmes with exotic dairy breeds for increasing milk production in West Africa.
Collapse
Affiliation(s)
- R C Mattioli
- International Trypanotolerance Centre, PMB 14, Banjul, Gambia.
| | | | | | | |
Collapse
|
29
|
Onah DN, Wakelin D. Trypanosome-induced suppression of responses to Trichinella spiralis in vaccinated mice. Int J Parasitol 1999; 29:1017-26. [PMID: 10501612 DOI: 10.1016/s0020-7519(99)00071-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mice vaccinated against the gastro-intestinal (GI) nematode Trichinella spiralis by injection of muscle larval homogenate antigen express a strong immunity to subsequent infection, reflected in earlier expulsion of adult worms from the intestine and reduced female worm fecundity. Infection with Trypanosoma brucei at the time of vaccination, or at the time of infection with T. spiralis, significantly reduced the level of immunity expressed, the effect being greatest when vaccination and T. brucei infection were given together. Trypanosome infection reduced T. spiralis-specific antibody responses in vaccinated mice, the effect being most apparent against IgM, IgG1 and IgG2b, and ablated the eosinophil response to T. spiralis. In vaccinated mice infected with both trypanosomes and T. spiralis, the proliferative responses of lymphocytes to the mitogen Con A or to T. spiralis antigen were much lower than in vaccinated mice infected only with the nematode. Whereas cells from mice infected only with T. spiralis produced the cytokine IL-4 and little or no IFNgamma when stimulated in vitro, cells from animals infected with T. spiralis and with trypanosomes released large amounts of IFNgamma but no IL-4. These observations are consistent with the known, IFNgamma-dependent, nitric-oxide-mediated suppressive effects of trypanosomes on lymphocyte function and the Th1 bias associated with these infections, both of which reduce the effectiveness of the Th2-mediated responses involved in immunity against GI nematode infections. The data are discussed in the context of the possible use of vaccines against GI nematodes in ruminants in countries where concurrent trypanosome-GI nematode infections are widespread.
Collapse
Affiliation(s)
- D N Onah
- School of Biological Sciences, University of Nottingham, UK
| | | |
Collapse
|
30
|
Buza J, Naessens J. Trypanosome non-specific IgM antibodies detected in serum of Trypanosoma congolense-infected cattle are polyreactive. Vet Immunol Immunopathol 1999; 69:1-9. [PMID: 10490230 DOI: 10.1016/s0165-2427(99)00033-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Serum Ig from Trypanosoma congolense-infected cattle were affinity-purified using immobilised trypanosome or non-trypanosome antigens (beta-galactosidase, cytochrome C and ferritin). The bound and unbound IgG and IgM fractions were collected and tested in ELISA for reactivity to each antigen. The results indicated that the presence of reactivity to non-parasite antigens in serum of infected cattle is due to polyreactive IgM antibodies. However, the IgG fraction only bound to trypanosome antigens and was only present in post-infection sera, indicating that it was induced by the infecting trypanosomes. Since the polyreactive IgM antibodies were also present in pre-infection sera, it is probable that they were natural antibodies that were not induced but only amplified by the trypanosome infection.
Collapse
Affiliation(s)
- J Buza
- International Livestock Research Institute, Nairobi, Kenya
| | | |
Collapse
|
31
|
|
32
|
Onah DN, Hopkins J, Luckins AG. Proliferative responses of peripheral blood leucocytes of sheep infected with Trypanosoma evansi. Scand J Immunol 1998; 48:170-6. [PMID: 9716108 DOI: 10.1046/j.1365-3083.1998.00354.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The effects of Trypanosoma evansi on the proliferative responses of ovine peripheral blood leucocytes (PBL) were examined in in vitro cell culture systems. Sheep were vaccinated against pneumonic pasteurellosis with a monovalent Pasteurella haemolytica vaccine and then infected with T. evansi TREU 2143. From 1 week post-infection (p.i.), the PBL were separated and stimulated in cultures with either Concanavalin A (Con A), bacterial lipopolysaccharide (LPS), pasteurella antigen (P.ag), or homologous trypanosome antigen (T.ag). The proliferative responses of the cells to Con A and LPS were significantly (P < 0.001) suppressed by the infection. This suppression was associated with active infection, as treatment of the sheep with a trypanocide restored the proliferative ability of the cells to both mitogens. Similarly, active infection significantly (P < 0.001) suppressed specific responses to P.ag and T.ag but although treatment resulted in full specific proliferative responsiveness to the homologous trypanosome antigen, the same was not true of P.ag, in which the responsiveness of cells from uninfected vaccinated sheep to it were still significantly higher (P < 0.001) than those of cells from infected sheep.
Collapse
Affiliation(s)
- D N Onah
- Centre for Tropical Veterinary Medicine, University of Edinburgh, UK
| | | | | |
Collapse
|
33
|
Buza J, Sileghem M, Gwakisa P, Naessens J. CD5+ B lymphocytes are the main source of antibodies reactive with non-parasite antigens in Trypanosoma congolense-infected cattle. Immunology 1997; 92:226-33. [PMID: 9415031 PMCID: PMC1364063 DOI: 10.1046/j.1365-2567.1997.00330.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Mice infected with African trypanosomes produce exceptionally large amounts of serum IgM, a major part of which binds to non-trypanosome antigens such as trinitrophenol and single-strand DNA. In this paper, we describe that in cattle infected with Trypanosoma congolense and T. vivax, similar antibodies are found, although they bind mainly to protein antigens, such as beta-galactosidase, ovalbumin and ferritin. The parasite non-specific IgM antibodies appear around the same time as the parasite-specific antibodies, but their origin and function are not clear. We tested the hypothesis that CD5+ B cells (or B-1 cells), which increase during trypanosome infections in cattle, are responsible for production of antibodies to non-trypanosome antigens. Splenic CD5+ and CD5- B cells from infected cattle were sorted and tested in a single cell blot assay. The numbers of immunoglobulin-secreting cells were similar in both B-cell populations. However, antibodies with reactivity for non-trypanosome antigens were significantly more prevalent in the CD5+ B-cell fraction and were exclusively IgM. The preference for production of these antibodies by CD5+ B cells and the expansion of this subpopulation during infections in cattle, strongly suggest that CD5+ B cells are the main source of trypanosome non-specific antibodies. We propose that these antibodies are natural, polyreactive antibodies that are predominantly secreted by CD5+ B cells. Since B-1 cells are up-regulated in many states of immune insufficiency, the immunosuppression associated with trypanosome infections may be responsible for the increase of this subset and the concomitant increase in trypanosome non-specific antibodies.
Collapse
Affiliation(s)
- J Buza
- International Livestock Research Institute, Nairobi, Kenya
| | | | | | | |
Collapse
|
34
|
Dwinger RH, Agyemang K, Kaufmann J, Grieve AS, Bah ML. Effects of trypanosome and helminth infections on health and production parameters of village N'Dama cattle in The Gambia. Vet Parasitol 1994; 54:353-65. [PMID: 7839560 DOI: 10.1016/0304-4017(94)90002-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The effects of trypanosome and helminth infections on health and production parameters in 2000 village N'Dama cattle were assessed periodically. Blood examination showed Trypanosoma congolense and Trypanosoma vivax to be prevalent, while strongylid-type eggs were those most frequently encountered in faecal samples. A distinct seasonal fluctuation was detected for both blood levels of trypanosomes and helminth egg output. Strongylid burden and trypanosome infection had significant negative effects on packed red cell volume levels and body weights mainly in animals of 2-3 years old. Clear indications of an increased susceptibility to trypanosomosis were found in animals affected by helminths. Similarly, animals infected with trypanosomes were more frequently infested with strongyles and egg counts were higher than in cattle in which no trypanosomes were detected.
Collapse
Affiliation(s)
- R H Dwinger
- International Trypanotolerance Centre (I.T.C.), Banjul, Gambia
| | | | | | | | | |
Collapse
|
35
|
Abstract
Recent biological investigations of the African trypanosomes have been moving away from their previous preoccupation with the phenomenon of antigenic variation. The feeling has arisen that antigenic variation, as demonstrated by the Trypanozoon and Nannomonas subgenera of trypanosomes, is too extensive, the number of serodemes too large and the coexistence of different species in many areas too complicated, to allow any immunoprophylaxis based on antibodies to variable antigens. This is, of course, not to rule out possible biochemical intervention in the biosynthesis or export of VSG molecules by trypanosomes. However, in the case of T. vivax, more information is required concerning antigenic variation and coat structure in this organism before these avenues of investigation are discarded. Ways of improving the yield of mature metacyclic trypanosomes in vitro must be found, so that the contribution of metacyclic variable antigens to the induction of immunity in T. vivax infection can be elucidated. The number of bloodstream VATs must be determined (perhaps by genetic rather than serological means), as there is evidence both for VAT exhaustion contributing to the self-cure of infected hosts, and for a possible limit to the number of VATs which can be expressed in infections in Africa. In South America nothing is known of the number of serodemes of T. vivax which exist, although such knowledge is obviously required, especially if immunity to bloodstream variants is the more important mechanism of inducing immunity to this trypanosome and true cyclical transmission is rare in, or absent from, that subcontinent. Further, in a fragile organism, with a coat of suspect integrity, the method of VSG packing and the relative exposure of underlying surface molecules seems to hold out even more hope for an immunological intervention based on cell surface but invariant molecules than is the case with T. brucei or T. congolense, although this is being attempted with the latter species. In T. brucei infections the appearance of the non-dividing stumpy population acts as a stimulus to the induction of humoral immune responses. In ruminants, antibody responses to T. vivax, at least as judged from lysis tests, lag behind the appearance of the different VATs by some days. It would be important to determine, therefore, whether, if late bloodstream forms could be induced more frequently in the ruminant, the speed of anti-VAT responses could be enhanced. Whilst self-cure appears to be relatively common in T. vivax infections, it is unlikely that it results in sterile immunity.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- P R Gardiner
- International Laboratory for Research on Animal Diseases, Nairobi, Kenya
| |
Collapse
|
36
|
|
37
|
Taracha EL, Irvin AD, Morzaria SP, Moloo SK, Katende JM, Kiarie JN. Effect of chronic trypanosomiasis on immunization against East Coast fever. Vet Parasitol 1986; 22:215-22. [PMID: 3564324 DOI: 10.1016/0304-4017(86)90108-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Two experiments were carried out in which uninfected cattle, or cattle chronically infected with Trypanosoma congolense, were immunized by the infection and treatment method against East Coast fever (ECF; Theileria parva infection). Chronic trypanosomiasis did not prevent cattle mounting an effective immunological response to ECF immunization and resisting subsequent lethal challenge. There appeared to be no difference in the level or quality of immunity between uninfected cattle and trypanosome-infected cattle. Thus, T. congolense infection on its own does not appear to provide a constraint to ECF immunization in the field.
Collapse
|
38
|
Askonas BA, Bancroft GJ. Interaction of African trypanosomes with the immune system. Philos Trans R Soc Lond B Biol Sci 1984; 307:41-9. [PMID: 6151687 DOI: 10.1098/rstb.1984.0107] [Citation(s) in RCA: 30] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
African trypanosomes cause disease in man and domestic animals. The parasites have the ability to escape immune control by two means: by antigenic variation of the surface glycoprotein coat so that waves of variant parasites arise and by inducing a general immunosuppression affecting immune responses to the parasite as well as to parasite-unrelated antigens. The cellular basis of the immune dysfunction will be discussed in relation to a mouse model system - it is the result of proliferative stimuli to T- or B-cells which then become refractory to selection by antigen and normal control signals. Recent experiments have focused on macrophages as important direct target cells for parasite action. We have obtained no evidence for a parasite derived mitogen acting directly on B- or T-cells. In vitro cell proliferation is associated with accessory cells and relates only to T-cells. During infection, macrophages become activated with changes in receptor expression and mediator release, so that there is, for example, spontaneous IL-1 release (with a role in T- and possibly in B-cell proliferation) and several-fold increases in PGE2 secretion, with its immunosuppressive activities. We also find parasitaemia-associated release of alpha-beta and gamma interferon by various cells which in turn influences immune function. The active parasite component is associated with parasite membranes, but its nature has not been further defined. We proposed that the macrophage changes provide a general pathway causing immune dysfunction associated with many infections, be they parasitic or caused by other invading organisms.
Collapse
|
39
|
Njau BC, Nyindo M, Mutani A. The interaction between the immune response of rabbits to heterologous antigens and a primary infestation with Rhipicephalus evertsi evertsi. Vet Res Commun 1990; 14:113-22. [PMID: 2345952 DOI: 10.1007/bf00346551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Rhipicephalus evertsi evertsi feeding on hosts inoculated with sheep red blood cells (SRBC) and bovine serum albumin (BSA) suppressed the primary antibody response to the two antigens. In addition, while the ticks paralysed most hosts in the studies, fatality associated with this toxicosis occurred only in rabbits which had received SRBC, either alone or with BSA. Only those hosts inoculated with BSA developed any resistance against the ticks, manifested by a slight reduction of engorged weights and development of anti-tick antibodies. These results suggest that R. e. evertsi infestation induces a degree of reduced host immune responsiveness to heterologous antigens.
Collapse
Affiliation(s)
- B C Njau
- International Centre of Insect Physiology and Ecology (ICIPE), Nairobi, Kenya
| | | | | |
Collapse
|