1
|
Siddiki AZ, Alam S, Fuad Bin Hossen F, Alim MA. Development of a multi-epitope chimeric vaccine in silico against Babesia bovis, Theileria annulata, and Anaplasma marginale using computational biology tools and reverse vaccinology approach. PLoS One 2025; 20:e0312262. [PMID: 39854345 PMCID: PMC11759392 DOI: 10.1371/journal.pone.0312262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/04/2024] [Indexed: 01/26/2025] Open
Abstract
The three rickettsial parasites- Babesia bovis, Theileria annulata and Anaplasma Marginale are responsible for causing Babesiosis, Theileriosis and Anaplasmosis among cattle. These diseases exist due to spreading of infected ticks. A large number of cattle were found to suffer from mixed infections caused by the three parasites at the same time. Due to these reasons cattle have been devoid of milk production with reduced meat availability. Hence, it is a matter of urgency for the immunity of cattle to exhibit resilience against all three rickettsial parasites. It could be possible if trials are carried out after producing a subunit chimeric vaccine against the rickettsial protozoan parasites and introducing it into the bloodstream of the cattle species. In this paper, we have used the process of reverse vaccinology to conduct a study in which we have developed a multi-epitope subunit chimeric vaccine against three protozoan parasites. We constructed three chimeric vaccine sequences from which only one chimeric vaccine construct was found to be an effective and efficient vaccine which is stable with high solubility and negative allergenicity. Following that, we performed molecular docking of the refined chimeric vaccine construct with Rp-105 and TLR-9. It was observed that the chimeric vaccines interacted with the receptors with high binding energy. Immune simulation was also performed to determine the potentiality of the chimeric vaccine for eliciting an immune response. The best-designed chimeric vaccine construct was then reverse transcribed and adapted for the host E. coli K12 strain which was later inserted into the pET28a (+) vector for the cloning and expression of the vaccine. The study could be a good initiative for the development of an effective chimeric vaccine against bovine parasites.
Collapse
Affiliation(s)
- Amam Zonaed Siddiki
- Department of Pathology and Parasitology, Chittagong Veterinary and Animal Sciences University (CVASU), Chittagong, Bangladesh
| | - Sabreena Alam
- Department of Biochemistry and Biotechnology, University of Science and Technology Chittagong (USTC), Chittagong, Bangladesh
| | - Farhan Fuad Bin Hossen
- Department of Biochemistry and Biotechnology, University of Science and Technology Chittagong (USTC), Chittagong, Bangladesh
| | - Md. Abdul Alim
- Department of Pathology and Parasitology, Chittagong Veterinary and Animal Sciences University (CVASU), Chittagong, Bangladesh
| |
Collapse
|
2
|
El-Alfy ES, Abbas I, Elseadawy R, El-Sayed SAES, Rizk MA. Genetic Diversity of Merozoite Surface Antigens in Global Babesia bovis Populations. Genes (Basel) 2023; 14:1936. [PMID: 37895285 PMCID: PMC10606690 DOI: 10.3390/genes14101936] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/05/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Cattle can be severely infected with the tick-borne protozoa Babesia bovis, giving rise to serious economic losses. Invasion of the host's RBCs by the parasite merozoite/sporozoites depends largely on the MSA (merozoite surface antigens) gene family, which comprises various fragments, e.g., MSA-1, MSA-2a1, MSA-2a2, MSA-2b and MSA-2c, highlighting the importance of these antigens as vaccine candidates. However, experimental trials documented the failure of some developed MSA-based vaccines to fully protect animals from B. bovis infection. One reason for this failure may be related to the genetic structure of the parasite. In the present study, all MSA-sequenced B. bovis isolates on the GenBank were collected and subjected to various analyses to evaluate their genetic diversity and population structure. The analyses were conducted on 199 MSA-1, 24 MSA-2a1, 193 MSA-2b and 148 MSA-2c isolates from geographically diverse regions. All these fragments displayed high nucleotide and haplotype diversities, but the MSA-1 was the most hypervariable and had the lowest inter- and intra-population gene flow values. This fragment also displayed a strong positive selection when testing its isolates for the natural selection, which suggests the potential occurrence of more genetic variations. On the contrary, the MSA-2c was the most conserved in comparison to the other fragments, and displayed the highest inter- and intra-population gene flow values, which was evidenced by a significantly negative selection and negative neutrality indices (Fu's Fs and Tajima's D). The majority of the MSA-2c tested isolates had two conserved amino acid repeats, and earlier reports have found these repeats to be highly immunogenic, which underlines the importance of this fragment in developing vaccines against B. bovis. Results of the MSA-2a1 analyses were also promising, but many more MSA-2a1 sequenced isolates are required to validating this assumption. The genetic analyses conducted for the MSA-2b fragment displayed borderline values when compared to the other fragments.
Collapse
Affiliation(s)
- El-Sayed El-Alfy
- Parasitology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt; (E.-S.E.-A.); (I.A.); (R.E.)
| | - Ibrahim Abbas
- Parasitology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt; (E.-S.E.-A.); (I.A.); (R.E.)
| | - Rana Elseadawy
- Parasitology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt; (E.-S.E.-A.); (I.A.); (R.E.)
| | - Shimaa Abd El-Salam El-Sayed
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Mohamed Abdo Rizk
- Department of Internal Medicine, Infectious and Fish Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
3
|
Mosqueda J, Hernandez-Silva DJ, Ueti MW, Cruz-Reséndiz A, Marquez-Cervantez R, Valdez-Espinoza UM, Dang-Trinh MA, Nguyen TT, Camacho-Nuez M, Mercado-Uriostegui MA, Aguilar-Tipacamú G, Ramos-Aragon JA, Hernandez-Ortiz R, Kawazu SI, Igarashi I. Spherical Body Protein 4 from Babesia bigemina: A Novel Gene That Contains Conserved B-Cell Epitopes and Induces Cross-Reactive Neutralizing Antibodies in Babesia ovata. Pathogens 2023; 12:pathogens12030495. [PMID: 36986418 PMCID: PMC10051436 DOI: 10.3390/pathogens12030495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/18/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Bovine babesiosis is a tick-transmitted disease caused by intraerythrocytic protozoan parasites of the genus Babesia. Its main causative agents in the Americas are Babesia bigemina and Babesia bovis, while Babesia ovata affects cattle in Asia. All Babesia species secrete proteins stored in organelles of the apical complex, which are involved in all steps of the invasion process of vertebrate host cells. Unlike other apicomplexans, which have dense granules, babesia parasites instead have large, round intracellular organelles called spherical bodies. Evidence suggests that proteins from these organelles are released during the process of invading red blood cells, where spherical body proteins (SBPs) play an important role in cytoskeleton reorganization. In this study, we characterized the gene that encodes SBP4 in B. bigemina. This gene is transcribed and expressed in the erythrocytic stages of B. bigemina. The sbp4 gene consists of 834 nucleotides without introns that encode a protein of 277 amino acids. In silico analysis predicted a signal peptide that is cleaved at residue 20, producing a 28.88-kDa protein. The presence of a signal peptide and the absence of transmembrane domains suggest that this protein is secreted. Importantly, when cattle were immunized with recombinant B. bigemina SBP4, antibodies identified B. bigemina and B. ovata merozoites according to confocal microscopy observations and were able to neutralize parasite multiplication in vitro for both species. Four peptides with predicted B-cell epitopes were identified to be conserved in 17 different isolates from six countries. Compared with the pre-immunization sera, antibodies against these conserved peptides reduced parasite invasion in vitro by 57%, 44%, 42%, and 38% for peptides 1, 2, 3, and 4, respectively (p < 0.05). Moreover, sera from cattle infected with B. bigemina cattle contained antibodies that recognized the individual peptides. All these results support the concept of spb4 as a new gene in B. bigemina that should be considered a candidate for a vaccine to control bovine babesiosis.
Collapse
Affiliation(s)
- Juan Mosqueda
- Immunology and Vaccines Laboratory, C. A. Facultad de Ciencias Naturales, Universidad Autonoma de Queretaro, Carretera a Chichimequillas, Ejido Bolaños, Queretaro 76140, Mexico
- C.A. Salud Animal y Microbiologia Ambiental, Facultad de Ciencias Naturales, Universidad Autonoma de Queretaro, Av. de las Ciencias s/n Col Juriquilla, Queretaro 76230, Mexico
| | - Diego Josimar Hernandez-Silva
- Immunology and Vaccines Laboratory, C. A. Facultad de Ciencias Naturales, Universidad Autonoma de Queretaro, Carretera a Chichimequillas, Ejido Bolaños, Queretaro 76140, Mexico
- Ph.D. Program in Biological Sciences, College of Natural Sciences, Autonomous University of Queretaro, Av. de las Ciencias s/n Col Juriquilla, Queretaro 76230, Mexico
| | - Massaro W Ueti
- Animal Diseases Research Unit, Agricultural Research Service, US Department of Agriculture, Pullman, WA 99164, USA
| | - Adolfo Cruz-Reséndiz
- Immunology and Vaccines Laboratory, C. A. Facultad de Ciencias Naturales, Universidad Autonoma de Queretaro, Carretera a Chichimequillas, Ejido Bolaños, Queretaro 76140, Mexico
- Veterinary Medicine Program, College of Natural Sciences, Autonomous University of Queretaro, Av. de las Ciencias s/n Col Juriquilla, Queretaro 76230, Mexico
| | - Ricardo Marquez-Cervantez
- Immunology and Vaccines Laboratory, C. A. Facultad de Ciencias Naturales, Universidad Autonoma de Queretaro, Carretera a Chichimequillas, Ejido Bolaños, Queretaro 76140, Mexico
- Veterinary Medicine Program, College of Natural Sciences, Autonomous University of Queretaro, Av. de las Ciencias s/n Col Juriquilla, Queretaro 76230, Mexico
| | - Uriel Mauricio Valdez-Espinoza
- Immunology and Vaccines Laboratory, C. A. Facultad de Ciencias Naturales, Universidad Autonoma de Queretaro, Carretera a Chichimequillas, Ejido Bolaños, Queretaro 76140, Mexico
- Master's Program in Animal Health and Production, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autonoma de Mexico, Av. Universidad 3000, Edificio A, Delegacion Coyoacan, Col. Ciudad Universitaria, Mexico City 04510, Mexico
- CENID-Salud Animal e Inocuidad/INIFAP, Carretera Federal Cuernavaca-Cuautla #8534, Col. Progreso, Jiutepec 62574, Mexico
| | - Minh-Anh Dang-Trinh
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, Japan
| | - Thu-Thuy Nguyen
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, Japan
| | - Minerva Camacho-Nuez
- Posgrado en Ciencias Genomicas, Universidad Autonoma de la Ciudad de Mexico, San Lorenzo 290, Esquina Roberto Gayol, Col. del Valle Sur, Delegacion Benito Juarez, Mexico City 03100, Mexico
| | - Miguel Angel Mercado-Uriostegui
- Immunology and Vaccines Laboratory, C. A. Facultad de Ciencias Naturales, Universidad Autonoma de Queretaro, Carretera a Chichimequillas, Ejido Bolaños, Queretaro 76140, Mexico
- C.A. Salud Animal y Microbiologia Ambiental, Facultad de Ciencias Naturales, Universidad Autonoma de Queretaro, Av. de las Ciencias s/n Col Juriquilla, Queretaro 76230, Mexico
| | - Gabriela Aguilar-Tipacamú
- C.A. Salud Animal y Microbiologia Ambiental, Facultad de Ciencias Naturales, Universidad Autonoma de Queretaro, Av. de las Ciencias s/n Col Juriquilla, Queretaro 76230, Mexico
| | - Juan Alberto Ramos-Aragon
- CENID-Salud Animal e Inocuidad/INIFAP, Carretera Federal Cuernavaca-Cuautla #8534, Col. Progreso, Jiutepec 62574, Mexico
| | - Ruben Hernandez-Ortiz
- CENID-Salud Animal e Inocuidad/INIFAP, Carretera Federal Cuernavaca-Cuautla #8534, Col. Progreso, Jiutepec 62574, Mexico
| | - Shin-Ichiro Kawazu
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, Japan
| | - Ikuo Igarashi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro 080-8555, Japan
| |
Collapse
|
4
|
Cuy-Chaparro L, Ricaurte-Contreras LA, Bohórquez MD, Arévalo-Pinzón G, Barreto-Santamaria A, Pabón L, Reyes C, Moreno-Pérez DA, Patarroyo MA. Identification of Babesia bovis MSA-1 functionally constraint regions capable of binding to bovine erythrocytes. Vet Parasitol 2022; 312:109834. [DOI: 10.1016/j.vetpar.2022.109834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/23/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
|
5
|
Transmission Cycle of Tick-Borne Infections and Co-Infections, Animal Models and Diseases. Pathogens 2022; 11:pathogens11111309. [PMID: 36365060 PMCID: PMC9696261 DOI: 10.3390/pathogens11111309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
Tick-borne pathogens such as species of Borrelia, Babesia, Anaplasma, Rickettsia, and Ehrlichia are widespread in the United States and Europe among wildlife, in passerines as well as in domestic and farm animals. Transmission of these pathogens occurs by infected ticks during their blood meal, carnivorism, and through animal bites in wildlife, whereas humans can become infected either by an infected tick bite, through blood transfusion and in some cases, congenitally. The reservoir hosts play an important role in maintaining pathogens in nature and facilitate transmission of individual pathogens or of multiple pathogens simultaneously to humans through ticks. Tick-borne co-infections were first reported in the 1980s in white-footed mice, the most prominent reservoir host for causative organisms in the United States, and they are becoming a major concern for public health now. Various animal infection models have been used extensively to better understand pathogenesis of tick-borne pathogens and to reveal the interaction among pathogens co-existing in the same host. In this review, we focus on the prevalence of these pathogens in different reservoir hosts, animal models used to investigate their pathogenesis and host responses they trigger to understand diseases in humans. We also documented the prevalence of these pathogens as correlating with the infected ticks’ surveillance studies. The association of tick-borne co-infections with other topics such as pathogens virulence factors, host immune responses as they relate to diseases severity, identification of vaccine candidates, and disease economic impact are also briefly addressed here.
Collapse
|
6
|
Babesia bovis AMA-1, MSA-2c and RAP-1 contain conserved B and T-cell epitopes, which generate neutralizing antibodies and a long-lasting Th1 immune response in vaccinated cattle. Vaccine 2022; 40:1108-1115. [DOI: 10.1016/j.vaccine.2022.01.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 02/02/2023]
|
7
|
Plasmepsin-like Aspartyl Proteases in Babesia. Pathogens 2021; 10:pathogens10101241. [PMID: 34684190 PMCID: PMC8540915 DOI: 10.3390/pathogens10101241] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/17/2021] [Accepted: 09/22/2021] [Indexed: 12/30/2022] Open
Abstract
Apicomplexan genomes encode multiple pepsin-family aspartyl proteases (APs) that phylogenetically cluster to six independent clades (A to F). Such diversification has been powered by the function-driven evolution of the ancestral apicomplexan AP gene and is associated with the adaptation of various apicomplexan species to different strategies of host infection and transmission through various invertebrate vectors. To estimate the potential roles of Babesia APs, we performed qRT-PCR-based expressional profiling of Babesia microti APs (BmASP2, 3, 5, 6), which revealed the dynamically changing mRNA levels and indicated the specific roles of individual BmASP isoenzymes throughout the life cycle of this parasite. To expand on the current knowledge on piroplasmid APs, we searched the EuPathDB and NCBI GenBank databases to identify and phylogenetically analyse the complete sets of APs encoded by the genomes of selected Babesia and Theileria species. Our results clearly determine the potential roles of identified APs by their phylogenetic relation to their homologues of known function—Plasmodium falciparum plasmepsins (PfPM I–X) and Toxoplasma gondii aspartyl proteases (TgASP1–7). Due to the analogies with plasmodial plasmepsins, piroplasmid APs represent valuable enzymatic targets that are druggable by small molecule inhibitors—candidate molecules for the yet-missing specific therapy for babesiosis.
Collapse
|
8
|
Elsworth B, Duraisingh MT. A framework for signaling throughout the life cycle of Babesia species. Mol Microbiol 2020; 115:882-890. [PMID: 33274587 DOI: 10.1111/mmi.14650] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 01/28/2023]
Abstract
Babesia species are tick-borne intracellular parasites that infect the red blood cells of their mammalian host, leading to severe or fatal disease. Babesia spp. infect a wide range of mammalian species and cause a significant economic burden globally, predominantly through disease in cattle. Several Babesia spp. are increasingly being recognized as zoonotic pathogens of humans. Babesia spp. have complex life cycles involving multiple stages in the tick and the mammalian host. The parasite utilizes complex signaling pathways during replication, egress, and invasion in each of these stages. They must also rapidly respond to their environment when switching between the mammalian and tick stages. This review will focus on the signaling pathways and environmental stimuli that Babesia spp. utilize in the bloodstream and for transmission to the tick, with an emphasis on the role of phosphorylation- and calcium-based signaling during egress and invasion. The expanding availability of in vitro and in vivo culture systems, genomes, transcriptomes, and transgenic systems available for a range of Babesia spp. should encourage further biological and translational studies of these ubiquitous parasites.
Collapse
Affiliation(s)
- Brendan Elsworth
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Manoj T Duraisingh
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
9
|
Matos CA, Silva JBD, Gonçalves LR, Mendes NS, Alvarez DO, André MR, Machado RZ. Genetic diversity of Babesia bovis studied longitudinally under natural transmission conditions in calves in the state of Rio de Janeiro, Brazil. ACTA ACUST UNITED AC 2020; 29:e021220. [PMID: 33237196 DOI: 10.1590/s1984-29612020089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 09/08/2020] [Indexed: 11/22/2022]
Abstract
Serum and DNA samples from 15 naturally infected calves in Seropédica, Brazil, were obtained quarterly from birth to 12 months of age, in order to longitudinally evaluate their humoral immune response against Babesia bovis and the merozoite surface antigen diversity of B. bovis. Anti-B. bovis IgG antibodies were detected by an indirect fluorescent antibody test (IFAT) and enzyme-linked immunosorbent assay (ELISA). Using DNA amplification, sequencing and phylogenetic analysis, the genetic diversity of B. bovis was assessed based on the genes that encode merozoite surface antigens (MSA-1, MSA-2b and MSA-2c). The serological results demonstrated that up to six months of age, all the calves developed active immunity against B. bovis. Among the 75 DNA samples evaluated, 0, 3 and 5 sequences of the msa-1, msa-2b and msa-2c genes were obtained, respectively. The present study demonstrated that the msa-2b and msa-2c gene sequences amplified from blood DNA of B. bovis-positive calves were genetically diversified. These data emphasize the importance of conducting deeper studies on the genetic diversity of B. bovis in Brazil, in order to design diagnostic antigens and vaccines in the future.
Collapse
Affiliation(s)
- Carlos António Matos
- Departamento de Patologia, Reprodução e Saúde Única, Faculdade de Ciências Agrárias e Veterinárias, Universidade Estadual Paulista (UNESP), Jaboticabal, SP, Brasil.,Direcção de Ciências Animais, Maputo, Moçambique
| | - Jenevaldo Barbosa da Silva
- Departamento de Patologia, Reprodução e Saúde Única, Faculdade de Ciências Agrárias e Veterinárias, Universidade Estadual Paulista (UNESP), Jaboticabal, SP, Brasil
| | - Luiz Ricardo Gonçalves
- Departamento de Patologia, Reprodução e Saúde Única, Faculdade de Ciências Agrárias e Veterinárias, Universidade Estadual Paulista (UNESP), Jaboticabal, SP, Brasil
| | - Natalia Serra Mendes
- Departamento de Patologia, Reprodução e Saúde Única, Faculdade de Ciências Agrárias e Veterinárias, Universidade Estadual Paulista (UNESP), Jaboticabal, SP, Brasil
| | | | - Marcos Rogério André
- Departamento de Patologia, Reprodução e Saúde Única, Faculdade de Ciências Agrárias e Veterinárias, Universidade Estadual Paulista (UNESP), Jaboticabal, SP, Brasil
| | - Rosangela Zacarias Machado
- Departamento de Patologia, Reprodução e Saúde Única, Faculdade de Ciências Agrárias e Veterinárias, Universidade Estadual Paulista (UNESP), Jaboticabal, SP, Brasil
| |
Collapse
|
10
|
Ueti MW, Johnson WC, Kappmeyer LS, Herndon DR, Mousel MR, Reif KE, Taus NS, Ifeonu OO, Silva JC, Suarez CE, Brayton KA. Comparative analysis of gene expression between Babesia bovis blood stages and kinetes allowed by improved genome annotation. Int J Parasitol 2020; 51:123-136. [PMID: 33069745 DOI: 10.1016/j.ijpara.2020.08.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/29/2020] [Accepted: 08/04/2020] [Indexed: 12/23/2022]
Abstract
Throughout their life cycle, Babesia parasites alternate between a mammalian host, where they cause babesiosis, and the tick vector. Transition between hosts results in distinct environmental signals that influence patterns of gene expression, consistent with the morphological and functional changes operating in the parasites during their life stages. In addition, comparing differential patterns of gene expression among mammalian and tick parasite stages can provide clues for developing improved methods of control. Hereby, we upgraded the genome assembly of Babesia bovis, a bovine hemoparasite, closing a 139 kbp gap, and used RNA-Seq datasets derived from mammalian blood and tick kinete stages to update the genome annotation. Of the originally annotated genes, 1,254 required structural changes, and 326 new genes were identified, leading to a different predicted proteome compared to the original annotation. Next, the RNA-Seq data was used to identify B. bovis genes that were differentially expressed in the vertebrate and arthropod hosts. In blood stages, 28% of the genes were upregulated up to 300 fold, whereas 26% of the genes in kinetes, a tick stage, were upregulated up to >19,000 fold. We thus discovered differentially expressed genes that may play key biological roles and serve as suitable targets for the development of vaccines to control bovine babesiosis.
Collapse
Affiliation(s)
- Massaro W Ueti
- Animal Diseases Research Unit, USDA-ARS, Pullman, Washington, USA; Program in Vector-borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA; Paul G. Allen School for Global Animal Health, Washington State University, Pullman, Washington, USA.
| | | | | | - David R Herndon
- Animal Diseases Research Unit, USDA-ARS, Pullman, Washington, USA
| | - Michelle R Mousel
- Animal Diseases Research Unit, USDA-ARS, Pullman, Washington, USA; Paul G. Allen School for Global Animal Health, Washington State University, Pullman, Washington, USA
| | - Kathryn E Reif
- Animal Diseases Research Unit, USDA-ARS, Pullman, Washington, USA; Program in Vector-borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Naomi S Taus
- Animal Diseases Research Unit, USDA-ARS, Pullman, Washington, USA; Program in Vector-borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Olukemi O Ifeonu
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Joana C Silva
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Carlos E Suarez
- Animal Diseases Research Unit, USDA-ARS, Pullman, Washington, USA; Program in Vector-borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Kelly A Brayton
- Program in Vector-borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA; Paul G. Allen School for Global Animal Health, Washington State University, Pullman, Washington, USA.
| |
Collapse
|
11
|
Characterization of the variable merozoite surface antigen (VMSA) gene family of Babesia orientalis. Parasitol Res 2020; 119:3639-3648. [PMID: 32930858 DOI: 10.1007/s00436-020-06877-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 09/06/2020] [Indexed: 01/01/2023]
Abstract
Due to its wide presence in apicomplexan parasites as well as high polymorphism and antigenic diversity, the variable merozoite surface antigen (VMSA) family in Babesia sp. has attracted increasing attention of researchers. Here, all the reported VMSA genes of Babesia spp. were obtained from GenBank, and multiple alignments were performed by using conserved regions to blast the Babesia orientalis genome database (unpublished data). Five MSA genes (named MSA-2a1, MSA-2a2, MSA-2c1, MSA-1, and MSA-2c2, respectively) were identified, sequenced, and cloned from B. orientalis, which were shown to encode proteins with open reading frames ranging in size from 266 (MSA-2c1) to 317 (MSA-1) amino acids. All the five proteins contain an MSA-2c superfamily conserved domain, with an identical signal peptide and glycosyl phosphatidyl inositol (GPI)-anchor for each of them. The five proteins were also predicted to contain B cell epitopes, with only three for BoMSA-2c1, the smallest protein in the BoVMSA family, while at least six for each of the others. Notably, BoMSA-2a1 has 2 identical copies, a specific phenomenon only present in B. orientalis. This research has determined the MSA genes of B. orientalis and provides a genetic basis for further research of functional genes in B. orientalis.
Collapse
|
12
|
Wang J, Yang J, Gao S, Wang X, Sun H, Lv Z, Li Y, Liu A, Liu J, Luo J, Guan G, Yin H. Genetic Diversity of Babesia bovis MSA-1, MSA-2b and MSA-2c in China. Pathogens 2020; 9:pathogens9060473. [PMID: 32549363 PMCID: PMC7350327 DOI: 10.3390/pathogens9060473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/09/2020] [Accepted: 06/12/2020] [Indexed: 11/23/2022] Open
Abstract
The apicomplexan parasite Babesia bovis is a tick-borne intracellular hemoprotozoan parasite that is widespread across China. Genetic diversity is an important strategy used by parasites to escape the immune responses of their hosts. In our present study, 575 blood samples, collected from cattle in 10 provinces, were initially screened using a nested PCR (polymerase chain reaction) for detection of B. bovis infection. To perform genetic diversity analyses, positive samples were further amplified to obtain sequences of three B. bovis merozoite surface antigen genes (MSA-1, MSA-2b, MSA-2c). The results of the nested PCR approach showed that an average of 8.9% (51/575) of cattle were positive for B. bovis infection. Phylogenetic analyses of the predicted amino acid sequences revealed that unique antigen variants were formed only by Chinese isolates. Our findings provide vital information for understanding the genetic diversity of B. bovis in China.
Collapse
Affiliation(s)
- Jinming Wang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou 730046, China; (J.W.); (J.Y.); (S.G.); (X.W.); (H.S.); (Z.L.); (Y.L.); (A.L.); (J.L.); (J.L.)
| | - Jifei Yang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou 730046, China; (J.W.); (J.Y.); (S.G.); (X.W.); (H.S.); (Z.L.); (Y.L.); (A.L.); (J.L.); (J.L.)
| | - Shandian Gao
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou 730046, China; (J.W.); (J.Y.); (S.G.); (X.W.); (H.S.); (Z.L.); (Y.L.); (A.L.); (J.L.); (J.L.)
| | - Xiaoxing Wang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou 730046, China; (J.W.); (J.Y.); (S.G.); (X.W.); (H.S.); (Z.L.); (Y.L.); (A.L.); (J.L.); (J.L.)
| | - Hao Sun
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou 730046, China; (J.W.); (J.Y.); (S.G.); (X.W.); (H.S.); (Z.L.); (Y.L.); (A.L.); (J.L.); (J.L.)
| | - Zhaoyong Lv
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou 730046, China; (J.W.); (J.Y.); (S.G.); (X.W.); (H.S.); (Z.L.); (Y.L.); (A.L.); (J.L.); (J.L.)
| | - Youquan Li
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou 730046, China; (J.W.); (J.Y.); (S.G.); (X.W.); (H.S.); (Z.L.); (Y.L.); (A.L.); (J.L.); (J.L.)
| | - Aihong Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou 730046, China; (J.W.); (J.Y.); (S.G.); (X.W.); (H.S.); (Z.L.); (Y.L.); (A.L.); (J.L.); (J.L.)
| | - Junlong Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou 730046, China; (J.W.); (J.Y.); (S.G.); (X.W.); (H.S.); (Z.L.); (Y.L.); (A.L.); (J.L.); (J.L.)
| | - Jianxun Luo
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou 730046, China; (J.W.); (J.Y.); (S.G.); (X.W.); (H.S.); (Z.L.); (Y.L.); (A.L.); (J.L.); (J.L.)
| | - Guiquan Guan
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou 730046, China; (J.W.); (J.Y.); (S.G.); (X.W.); (H.S.); (Z.L.); (Y.L.); (A.L.); (J.L.); (J.L.)
- Correspondence: (G.G.); (H.Y.)
| | - Hong Yin
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou 730046, China; (J.W.); (J.Y.); (S.G.); (X.W.); (H.S.); (Z.L.); (Y.L.); (A.L.); (J.L.); (J.L.)
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonose, Yangzhou University, Yangzhou 225009, China
- Correspondence: (G.G.); (H.Y.)
| |
Collapse
|
13
|
Li M, Ao Y, Guo J, Nie Z, Liu Q, Yu L, Luo X, Zhan X, Zhao Y, Wang S, An X, He L, Zhao J. Surface Antigen 1 Is a Crucial Secreted Protein That Mediates Babesia microti Invasion Into Host Cells. Front Microbiol 2020; 10:3046. [PMID: 32010102 PMCID: PMC6974462 DOI: 10.3389/fmicb.2019.03046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 12/18/2019] [Indexed: 12/16/2022] Open
Abstract
Babesia microti, a tick-borne intraerythrocytic zoonotic protozoan, causes most of human babesiosis in the world, and patients usually experience intermittent fever, fatigue, and chills, followed by a combination of additional symptoms and even death in severe cases. Unfortunately, there is no curable drug or effective vaccine available, and the mechanism of related virulence factors in invasion to host cells during the merozoite stage is unclear. Here, we evaluated a secreted protein annotated as B. microti surface antigen 1 (BmSA1) and identified from in vitro culture supernatant by liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS). BmSA1 fragment was expressed in Escherichia coli to prepare polyclonal antiserum. Western blot analysis revealed the existence of BmSA1 in the lysate of the parasites and the hemolysate of infected red blood cells (iRBCs). Laser confocal microscopy confirmed BmSA1 as a secreted protein with diffuse distribution around the parasites in red blood cells (RBCs). The adhesion capacity of BmSA1 against the host RBCs was tested by RBC binding assays using the recombinant BmSA1 protein (rBmSA1), which was shown to specifically bind to host RBCs. Further in vitro antiserum-neutralization test demonstrated that the growth of parasites could be significantly inhibited by the anti-BmSA1 antiserum. These results indicate that BmSA1 is a crucial factor for B. microti invasion into host RBCs with an important role in host-parasite interactions during the merozoite stage and has the potential use as a vaccine candidate due to its high secretion amount.
Collapse
Affiliation(s)
- Muxiao Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yangsiqi Ao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Jiaying Guo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Zheng Nie
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Qin Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Long Yu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiaoying Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xueyan Zhan
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yangnan Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Sen Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiaomeng An
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Lan He
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Animal Epidemical Disease and Infectious Zoonoses, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Junlong Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Animal Epidemical Disease and Infectious Zoonoses, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
14
|
Babesiosis Vaccines: Lessons Learned, Challenges Ahead, and Future Glimpses. Trends Parasitol 2019; 35:622-635. [PMID: 31281025 DOI: 10.1016/j.pt.2019.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/06/2019] [Accepted: 06/06/2019] [Indexed: 01/02/2023]
Abstract
The incidence and prevalence of babesiosis in animals and humans is increasing, yet prevention, control, or treatment measures remain limited and ineffective. Despite a growing body of new knowledge of the biology, pathogenicity, and virulence of Babesia parasites, there is still no well-defined, adequately effective and easily deployable vaccine. While numerous published studies suggest that the development of such anti-Babesia vaccines should be feasible, many others identify significant challenges that need to be overcome in order to succeed. Here, we review historic and recent attempts in babesiosis vaccine discovery to avoid past pitfalls, learn new lessons, and provide a roadmap to guide the development of next-generation babesiosis vaccines.
Collapse
|
15
|
Matos CA, Gonçalves LR, Alvarez DO, Freschi CR, Silva JBD, Val-Moraes SP, Mendes NS, André MR, Machado RZ. Longitudinal evaluation of humoral immune response and merozoite surface antigen diversity in calves naturally infected with Babesia bovis, in São Paulo, Brazil. ACTA ACUST UNITED AC 2018; 26:479-490. [PMID: 29211135 DOI: 10.1590/s1984-29612017069] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 11/07/2017] [Indexed: 11/21/2022]
Abstract
Babesiosis is an economically important infectious disease affecting cattle worldwide. In order to longitudinally evaluate the humoral immune response against Babesia bovis and the merozoite surface antigen diversity of B. bovis among naturally infected calves in Taiaçu, Brazil, serum and DNA samples from 15 calves were obtained quarterly, from their birth to 12 months of age. Anti-B. bovis IgG antibodies were detected by means of the indirect fluorescent antibody test (IFAT) and enzyme-linked immunosorbent assay (ELISA). The polymerase chain reaction (PCR) was used to investigate the genetic diversity of B. bovis, based on the genes that encode merozoite surface antigens (MSA-1, MSA-2b and MSA-2c). The serological results demonstrated that up to six months of age, all the calves developed active immunity against B. bovis. Among the 75 DNA samples evaluated, 2, 4 and 5 sequences of the genes msa-1, msa-2b and msa-2c were obtained. The present study demonstrated that the msa-1 and msa-2b genes sequences amplified from blood DNA of calves positive to B. bovis from Taiaçu were genetically distinct, and that msa-2c was conserved. All animals were serologically positive to ELISA and IFAT, which used full repertoire of parasite antigens in despite of the genetic diversity of MSAs.
Collapse
Affiliation(s)
- Carlos António Matos
- Laboratório de Imunoparasitologia, Departamento de Patologia Veterinária, Universidade Estadual Paulista - UNESP, Jaboticabal, SP, Brasil.,Direcção de Ciências Animais, Maputo, Moçambique
| | - Luiz Ricardo Gonçalves
- Laboratório de Imunoparasitologia, Departamento de Patologia Veterinária, Universidade Estadual Paulista - UNESP, Jaboticabal, SP, Brasil
| | | | - Carla Roberta Freschi
- Laboratório de Imunoparasitologia, Departamento de Patologia Veterinária, Universidade Estadual Paulista - UNESP, Jaboticabal, SP, Brasil
| | - Jenevaldo Barbosa da Silva
- Laboratório de Imunoparasitologia, Departamento de Patologia Veterinária, Universidade Estadual Paulista - UNESP, Jaboticabal, SP, Brasil
| | - Silvana Pompeia Val-Moraes
- Laboratório de Imunoparasitologia, Departamento de Patologia Veterinária, Universidade Estadual Paulista - UNESP, Jaboticabal, SP, Brasil
| | - Natalia Serra Mendes
- Laboratório de Imunoparasitologia, Departamento de Patologia Veterinária, Universidade Estadual Paulista - UNESP, Jaboticabal, SP, Brasil
| | - Marcos Rogério André
- Laboratório de Imunoparasitologia, Departamento de Patologia Veterinária, Universidade Estadual Paulista - UNESP, Jaboticabal, SP, Brasil
| | - Rosangela Zacarias Machado
- Laboratório de Imunoparasitologia, Departamento de Patologia Veterinária, Universidade Estadual Paulista - UNESP, Jaboticabal, SP, Brasil
| |
Collapse
|
16
|
Man S, Fu Y, Guan Y, Feng M, Qiao K, Li X, Gao H, Cheng X. Evaluation of a Major Surface Antigen of Babesia microti Merozoites as a Vaccine Candidate against Babesia Infection. Front Microbiol 2017; 8:2545. [PMID: 29312230 PMCID: PMC5742146 DOI: 10.3389/fmicb.2017.02545] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 12/07/2017] [Indexed: 02/02/2023] Open
Abstract
Babesia species are tick-borne intraerythrocytic protozoa that cause babesiosis in humans worldwide. No vaccine has yet proven effective against Babesia infection. Surface antigens of merozoites are involved in the invasion of erythrocytes by Babesia. Surface antigens may be presented by both babesial sporozoites and merozoites and provide a general target for antibody-mediated inhibition of erythrocyte invasion. Here we evaluated a major surface antigen of B. microti merozoites, BMSA, as a potential vaccine to prevent babesiosis. Our data indicated that bmsa is transcribed during different phases, including ring form, amoeboid form, and merozoites, and that its expression is significantly increased in mature merozoites. The protein was found to be located in the membrane of B. microti and in the cytoplasm of infected erythrocytes. The immune response induced by BMSA had a significant inhibitory effect on parasite invasion of the host erythrocytes (83.3% inhibition of invasion) and parasite growth in vivo. The levels of parasitemia significantly decreased after BMSA vaccination when mice were infected with babesia parasite. Importantly, protective immunity was significantly related to the upregulation of the Th17 cytokine interleukin-17, the Th1 cytokine interleukin-12p70 and the Th2 cytokines, such as interleukin-4, -6, and -10. Ingenuity Pathway Analysis indicated that interleukin-17 facilitated the secretion of Th2 cytokines, such as interleukin-10, -4, and -6, thereby inducing a predominately Th2 protective immune response and promoting the expression a high level of special IgG1 against Babesia infection. Further, an anti-BMSA monoclonal antibody successfully protected NOD/SCID mice from a challenge with B. microti. Taken together, our results indicated that BMSA induces a protective immune response against Babesia infection and may serve as a potential vaccine.
Collapse
Affiliation(s)
- Suqin Man
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yongfeng Fu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yue Guan
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Meng Feng
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ke Qiao
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xueping Li
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hongjian Gao
- Department of Electron Microscopy, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Xunjia Cheng
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Institute of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Johnson WC, Taus NS, Reif KE, Bohaliga GAR, Kappmeyer LS, Ueti MW. Analysis of Stage-Specific Protein Expression during Babesia Bovis Development within Female Rhipicephalus Microplus. J Proteome Res 2017; 16:1327-1338. [PMID: 28152313 DOI: 10.1021/acs.jproteome.6b00947] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Arthropod-borne protozoan pathogens have a complex life cycle that includes asexual reproduction of haploid stages in mammalian hosts and the development of diploid stages in invertebrate hosts. The ability of pathogens to invade, survive, and replicate within distinct cell types is required to maintain their life cycle. In this study, we describe a comparative proteomic analysis of a cattle pathogen, Babesia bovis, during its development within the mammalian and tick hosts with the goal of identifying cell-surface proteins expressed by B. bovis kinetes as potential targets for the development of a transmission blocking vaccine. To determine parasite tick-stage-specific cell-surface proteins, CyDye labeling was performed with B. bovis blood stages from the bovine host and kinetes from the tick vector. Cell-surface kinete-stage-specific proteins were identified using 2D difference in gel electrophoresis and analyzed by mass spectrometry. Ten proteins were identified as kinete-stage-specific, with orthologs found in closely related Apicomplexan pathogens. Transcriptional analysis revealed two genes were highly expressed by kinetes as compared with blood stages. Immunofluorescence using antibodies against the two proteins confirmed kinete-stage-specific expression. The identified cell-surface kinete proteins are potential candidates for the development of a B. bovis transmission blocking vaccine.
Collapse
Affiliation(s)
- Wendell C Johnson
- Animal Disease Research Unit, USDA-ARS , Pullman, Washington 99164, United States
| | - Naomi S Taus
- Animal Disease Research Unit, USDA-ARS , Pullman, Washington 99164, United States.,Program in Vector-borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State University , Pullman, Washington 99164, United States
| | - Kathryn E Reif
- Animal Disease Research Unit, USDA-ARS , Pullman, Washington 99164, United States.,Program in Vector-borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State University , Pullman, Washington 99164, United States
| | - Gamila A R Bohaliga
- Program in Vector-borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State University , Pullman, Washington 99164, United States
| | - Lowell S Kappmeyer
- Animal Disease Research Unit, USDA-ARS , Pullman, Washington 99164, United States
| | - Massaro W Ueti
- Animal Disease Research Unit, USDA-ARS , Pullman, Washington 99164, United States.,Program in Vector-borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State University , Pullman, Washington 99164, United States.,Paul G. Allen School for Global Animal Health, Washington State University , Pullman, Washington 99164, United States
| |
Collapse
|
18
|
Genetic diversity and antigenicity variation of Babesia bovis merozoite surface antigen-1 (MSA-1) in Thailand. INFECTION GENETICS AND EVOLUTION 2016; 41:255-261. [PMID: 27101782 DOI: 10.1016/j.meegid.2016.04.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 03/04/2016] [Accepted: 04/17/2016] [Indexed: 11/21/2022]
Abstract
Babesia bovis, an intraerythrocytic protozoan parasite, causes severe clinical disease in cattle worldwide. The genetic diversity of parasite antigens often results in different immune profiles in infected animals, hindering efforts to develop immune control methodologies against the B. bovis infection. In this study, we analyzed the genetic diversity of the merozoite surface antigen-1 (msa-1) gene using 162 B. bovis-positive blood DNA samples sourced from cattle populations reared in different geographical regions of Thailand. The identity scores shared among 93 msa-1 gene sequences isolated by PCR amplification were 43.5-100%, and the similarity values among the translated amino acid sequences were 42.8-100%. Of 23 total clades detected in our phylogenetic analysis, Thai msa-1 gene sequences occurred in 18 clades; seven among them were composed of sequences exclusively from Thailand. To investigate differential antigenicity of isolated MSA-1 proteins, we expressed and purified eight recombinant MSA-1 (rMSA-1) proteins, including an rMSA-1 from B. bovis Texas (T2Bo) strain and seven rMSA-1 proteins based on the Thai msa-1 sequences. When these antigens were analyzed in a western blot assay, anti-T2Bo cattle serum strongly reacted with the rMSA-1 from T2Bo, as well as with three other rMSA-1 proteins that shared 54.9-68.4% sequence similarity with T2Bo MSA-1. In contrast, no or weak reactivity was observed for the remaining rMSA-1 proteins, which shared low sequence similarity (35.0-39.7%) with T2Bo MSA-1. While demonstrating the high genetic diversity of the B. bovis msa-1 gene in Thailand, the present findings suggest that the genetic diversity results in antigenicity variations among the MSA-1 antigens of B. bovis in Thailand.
Collapse
|
19
|
Ishizaki T, Sivakumar T, Hayashida K, Tuvshintulga B, Igarashi I, Yokoyama N. RBC invasion and invasion-inhibition assays using free merozoites isolated after cold treatment of Babesia bovis in vitro culture. Exp Parasitol 2016; 166:10-5. [PMID: 26965399 DOI: 10.1016/j.exppara.2016.03.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 02/24/2016] [Accepted: 03/05/2016] [Indexed: 10/22/2022]
Abstract
Babesia bovis is an apicomplexan hemoprotozoan that can invade bovine red blood cells (RBCs), where it multiplies asexually. RBC invasion assays using free viable merozoites are now routinely used to understand the invasion mechanism of B. bovis, and to evaluate the efficacy of chemicals and antibodies that potentially inhibit RBC invasion by the parasite. The application of high-voltage pulses (high-voltage electroporation), a commonly used method to isolate free merozoites from infected RBCs, reduces the viability of the merozoites. Recently, a cold treatment of B. bovis in vitro culture was found to induce an effective release of merozoites from the infected RBCs. In the present study, we incubated in vitro cultures of B. bovis in an ice bath to liberate merozoites from infected RBCs and then evaluated the isolated merozoites in RBC invasion and invasion-inhibitions assays. The viability of the purified merozoites (72.4%) was significantly higher than that of merozoites isolated with high-voltage electroporation (48.5%). The viable merozoites prepared with the cold treatment also invaded uninfected bovine RBCs at a higher rate (0.572%) than did merozoites prepared with high-voltage electroporation (0.251%). The invasion-blocking capacities of heparin, a polyclonal rabbit antibody directed against recombinant B. bovis rhoptry associated protein 1, and B. bovis-infected bovine serum were successfully demonstrated in an RBC invasion assay with the live merozoites prepared with the cold treatment, suggesting that the targets of these inhibitors were intact in the merozoites. These findings indicate that the cold treatment technique is a useful tool for the isolation of free, viable, invasion-competent B. bovis merozoites, which can be effectively used for RBC invasion and invasion-inhibition assays in Babesia research.
Collapse
Affiliation(s)
- Takahiro Ishizaki
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | - Thillaiampalam Sivakumar
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | - Kyoko Hayashida
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | - Bumduuren Tuvshintulga
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | - Ikuo Igarashi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | - Naoaki Yokoyama
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan.
| |
Collapse
|
20
|
Liyanagunawardena N, Sivakumar T, Kothalawala H, Silva SSP, Battsetseg B, Lan DTB, Inoue N, Igarashi I, Yokoyama N. Type-specific PCR assays for Babesia bovis msa-1 genotypes in Asia: Revisiting the genetic diversity in Sri Lanka, Mongolia, and Vietnam. INFECTION GENETICS AND EVOLUTION 2015; 37:64-9. [PMID: 26520797 DOI: 10.1016/j.meegid.2015.10.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 10/15/2015] [Accepted: 10/27/2015] [Indexed: 11/17/2022]
Abstract
Babesia bovis is the most virulent Babesia organism, resulting in a high mortality rate in cattle. The genetic diversity of B. bovis merozoite surface antigens (MSAs), such as MSA-1, MSA-2b, and MSA-2c, might be linked to altered immune profiles in the host animals. The present study aimed to develop type-specific PCR assays for Asian msa-1 genotypes, thereby re-analyzing the genetic diversity of msa-1 in Sri Lanka, Mongolia, and Vietnam. Specific primers were designed for nine Asian msa-1 genotypes, which had been detected based on the phylogeny constructed using msa-1 gene sequences retrieved from the GenBank database. Specificity of the type-specific PCR assays was confirmed using plasmids containing the inserts of msa-1 gene fragments that represent Asian genotypes. Furthermore, no amplicons were observed by these PCR assays when DNA samples of Babesia bigemina, Babesia ovata, Theileria annulata, Theileria orientalis, Trypanosoma evansi, Trypanosoma theileri, Anaplasma marginale, and Anaplasma bovis, and non-infected bovine blood were analyzed. In total, 109 B. bovis-positive blood DNA samples sourced from Sri Lanka (44 cattle), Mongolia (26 cattle), and Vietnam (23 cattle and 16 water buffaloes) were then screened by the type-specific PCR assays. The sequences derived from all of the PCR amplicons were phylogenetically analyzed. Out of 109 DNA samples, 23 (20 from cattle and 3 from water buffaloes) were positive for at least one genotype. In agreement with previous studies, five and four different genotypes were detected among the DNA samples from Sri Lanka and Vietnam, respectively. In contrast, four genotypes, including three novel genotypes, were detected from Mongolia. Five DNA samples were found to be co-infected with multiple genotypes. The sequences of the PCR amplicons clustered phylogenetically within the corresponding clades. These findings indicated that the type-specific PCR assays described herein are useful for the determination of genotypic diversity of the B. bovis msa-1 gene in Asia.
Collapse
Affiliation(s)
- Nilukshi Liyanagunawardena
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan; Veterinary Research Institute, Peradeniya, Sri Lanka
| | - Thillaiampalam Sivakumar
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan; Veterinary Research Institute, Peradeniya, Sri Lanka
| | | | | | - Badgar Battsetseg
- Laboratory of Molecular Genetics, Institute of Veterinary Medicine, Ulaanbaatar, Mongolia
| | | | - Noboru Inoue
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Ikuo Igarashi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Naoaki Yokoyama
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan.
| |
Collapse
|
21
|
Florin-Christensen M, Suarez CE, Rodriguez AE, Flores DA, Schnittger L. Vaccines against bovine babesiosis: where we are now and possible roads ahead. Parasitology 2014; 141:1563-1592. [PMID: 25068315 DOI: 10.1017/s0031182014000961] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bovine babesiosis caused by the tick-transmitted haemoprotozoans Babesia bovis, Babesia bigemina and Babesia divergens commonly results in substantial cattle morbidity and mortality in vast world areas. Although existing live vaccines confer protection, they have considerable disadvantages. Therefore, particularly in countries where large numbers of cattle are at risk, important research is directed towards improved vaccination strategies. Here a comprehensive overview of currently used live vaccines and of the status quo of experimental vaccine trials is presented. In addition, pertinent research fields potentially contributing to the development of novel non-live and/or live vaccines are discussed, including parasite antigens involved in host cell invasion and in pathogen-tick interactions, as well as the protective immunity against infection. The mining of available parasite genomes is continuously enlarging the array of potential vaccine candidates and, additionally, the recent development of a transfection tool for Babesia can significantly contribute to vaccine design. However, the complication and high cost of vaccination trials hinder Babesia vaccine research, and have so far seriously limited the systematic examination of antigen candidates and prevented an in-depth testing of formulations using different immunomodulators and antigen delivery systems.
Collapse
Affiliation(s)
- Monica Florin-Christensen
- Instituto de Patobiologia, CICVyA, INTA-Castelar, 1686 Hurlingham, Argentina
- CONICET, C1033AAJ Ciudad Autonoma de Buenos Aires, Argentina
| | - Carlos E Suarez
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA 99164-7040, USA
- ADRU-ARS, United States Department of Agriculture, Pullman, WA 99164-6630, USA
| | - Anabel E Rodriguez
- Instituto de Patobiologia, CICVyA, INTA-Castelar, 1686 Hurlingham, Argentina
| | - Daniela A Flores
- Instituto de Patobiologia, CICVyA, INTA-Castelar, 1686 Hurlingham, Argentina
- ANPCyT, C1425FQD Ciudad Autonoma de Buenos Aires, Argentina
| | - Leonhard Schnittger
- Instituto de Patobiologia, CICVyA, INTA-Castelar, 1686 Hurlingham, Argentina
- CONICET, C1033AAJ Ciudad Autonoma de Buenos Aires, Argentina
| |
Collapse
|
22
|
Molad T, Fleiderovitz L, Leibovich B, Wolkomirsky R, Erster O, Roth A, Mazuz M, Markovics A, Shkap V. Genetic polymorphism of Babesia bovis merozoite surface antigens-2 (MSA-2) isolates from bovine blood and Rhipicephalus annulatus ticks in Israel. Vet Parasitol 2014; 205:20-7. [DOI: 10.1016/j.vetpar.2014.07.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 06/27/2014] [Accepted: 07/10/2014] [Indexed: 11/17/2022]
|
23
|
Rodriguez AE, Florin-Christensen M, Flores DA, Echaide I, Suarez CE, Schnittger L. The glycosylphosphatidylinositol-anchored protein repertoire of Babesia bovis and its significance for erythrocyte invasion. Ticks Tick Borne Dis 2014; 5:343-8. [DOI: 10.1016/j.ttbdis.2013.12.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 12/23/2013] [Accepted: 12/29/2013] [Indexed: 11/26/2022]
|
24
|
Tattiyapong M, Sivakumar T, Ybanez AP, Ybanez RHD, Perez ZO, Guswanto A, Igarashi I, Yokoyama N. Diversity of Babesia bovis merozoite surface antigen genes in the Philippines. Parasitol Int 2014; 63:57-63. [DOI: 10.1016/j.parint.2013.09.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 08/07/2013] [Accepted: 09/05/2013] [Indexed: 10/26/2022]
|
25
|
The molecular prevalence and MSA-2b gene-based genetic diversity of Babesia bovis in dairy cattle in Thailand. Vet Parasitol 2013; 197:642-8. [PMID: 23953761 DOI: 10.1016/j.vetpar.2013.07.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 07/08/2013] [Accepted: 07/09/2013] [Indexed: 11/20/2022]
Abstract
Bovine babesiosis is an economically significant disease that affects dairy farming operations in Thailand. In the present study, 1824 blood-DNA samples prepared from cattle bred in 4 different regions of the country (North, Northeast, Central, and South) were screened using a nested PCR for the specific detection of Babesia bovis. While the overall prevalence of B. bovis was 8.8%, the Central region of Thailand was found to be a high-risk area of the country, as the prevalence of the parasite was 15.0%. The positive rate was relatively higher among the animals of 1-5 years of age. The genetic diversity among the B. bovis parasites was also studied based on their MSA-2b gene, and the findings showed that the Thai sequences were dispersed across 8 of 13 total clades observed in the phylogram. Three of these clades were formed only of Thai sequences. Similarity among the deduced MSA-2b amino acid sequences determined in the present study was 68.3-100%. In conclusion, the present study found that all the locations surveyed were infected with B. bovis and that the parasite populations in Thailand were genetically diverse. Our findings highlight the need for further studies in Thailand to generate more information before a sound control strategy could be implemented against B. bovis.
Collapse
|
26
|
Nagano D, Sivakumar T, De De Macedo ACC, Inpankaew T, Alhassan A, Igarashi I, Yokoyama N. The genetic diversity of merozoite surface antigen 1 (MSA-1) among Babesia bovis detected from cattle populations in Thailand, Brazil and Ghana. J Vet Med Sci 2013; 75:1463-70. [PMID: 23856760 PMCID: PMC3942984 DOI: 10.1292/jvms.13-0251] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In the present study, we screened blood DNA samples obtained from cattle bred in Brazil
(n=164) and Ghana (n=80) for Babesia bovis using a diagnostic PCR assay
and found prevalences of 14.6% and 46.3%, respectively. Subsequently, the genetic
diversity of B. bovis in Thailand, Brazil and Ghana was analyzed, based
on the DNA sequence of merozoite surface antigen-1 (MSA-1). In Thailand,
MSA-1 sequences were relatively conserved and found in a single clade
of the phylogram, while Brazilian MSA-1 sequences showed high genetic
diversity and were dispersed across three different clades. In contrast, the sequences
from Ghanaian samples were detected in two different clades, one of which contained only a
single Ghanaian sequence. The identities among the MSA-1 sequences from
Thailand, Brazil and Ghana were 99.0–100%, 57.5–99.4% and 60.3–100%, respectively, while
the similarities among the deduced MSA-1 amino acid sequences within the respective
countries were 98.4–100%, 59.4–99.7% and 58.7–100%, respectively. These observations
suggested that the genetic diversity of B. bovis based on
MSA-1 sequences was higher in Brazil and Ghana than in Thailand. The
current data highlight the importance of conducting extensive studies on the genetic
diversity of B. bovis before designing immune control strategies in each
surveyed country.
Collapse
Affiliation(s)
- Daisuke Nagano
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Gohil S, Kats LM, Seemann T, Fernandez KM, Siddiqui G, Cooke BM. Bioinformatic prediction of the exportome of Babesia bovis and identification of novel proteins in parasite-infected red blood cells. Int J Parasitol 2013; 43:409-16. [PMID: 23395698 DOI: 10.1016/j.ijpara.2013.01.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 01/15/2013] [Accepted: 01/16/2013] [Indexed: 10/27/2022]
Abstract
Babesia bovis is a pathogen of considerable economic significance to the livestock industry worldwide but the precise mechanisms by which this parasite causes disease in susceptible cattle remain poorly understood. It is clear, however, that alterations to the structure and function of red blood cells in which the parasites reside and replicate play an important role in pathogenesis and that these are secondary to the export of numerous, currently unknown and uncharacterised parasite-encoded proteins. Using a rational bioinformatic approach, we have identified a set of 362 proteins (117 of which are hypothetical) that we predict encompasses the B. bovis exportome. These exported proteins are likely to be trafficked to various cellular locations, with a subset destined for the red blood cell cytosol or the red blood cell cytoskeleton. These proteins are likely to play important roles in mediating the pathogenesis of babesiosis. We have selected three novel proteins and confirmed their predicted export and localisation within the host red blood cell by immunofluorescence using specific antibodies raised against these proteins. Complete characterisation of these novel exported parasite proteins will help elucidate their function within the host red blood cell and assist in identification of new therapeutic targets for babesiosis.
Collapse
Affiliation(s)
- Sejal Gohil
- Department of Microbiology, Monash University, Victoria 3800, Australia
| | | | | | | | | | | |
Collapse
|
28
|
Esch KJ, Petersen CA. Transmission and epidemiology of zoonotic protozoal diseases of companion animals. Clin Microbiol Rev 2013; 26:58-85. [PMID: 23297259 PMCID: PMC3553666 DOI: 10.1128/cmr.00067-12] [Citation(s) in RCA: 179] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Over 77 million dogs and 93 million cats share our households in the United States. Multiple studies have demonstrated the importance of pets in their owners' physical and mental health. Given the large number of companion animals in the United States and the proximity and bond of these animals with their owners, understanding and preventing the diseases that these companions bring with them are of paramount importance. Zoonotic protozoal parasites, including toxoplasmosis, Chagas' disease, babesiosis, giardiasis, and leishmaniasis, can cause insidious infections, with asymptomatic animals being capable of transmitting disease. Giardia and Toxoplasma gondii, endemic to the United States, have high prevalences in companion animals. Leishmania and Trypanosoma cruzi are found regionally within the United States. These diseases have lower prevalences but are significant sources of human disease globally and are expanding their companion animal distribution. Thankfully, healthy individuals in the United States are protected by intact immune systems and bolstered by good nutrition, sanitation, and hygiene. Immunocompromised individuals, including the growing number of obese and/or diabetic people, are at a much higher risk of developing zoonoses. Awareness of these often neglected diseases in all health communities is important for protecting pets and owners. To provide this awareness, this review is focused on zoonotic protozoal mechanisms of virulence, epidemiology, and the transmission of pathogens of consequence to pet owners in the United States.
Collapse
|
29
|
Gohil S, Herrmann S, Günther S, Cooke BM. Bovine babesiosis in the 21st century: advances in biology and functional genomics. Int J Parasitol 2012; 43:125-32. [PMID: 23068911 DOI: 10.1016/j.ijpara.2012.09.008] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2012] [Revised: 09/21/2012] [Accepted: 09/24/2012] [Indexed: 10/27/2022]
Abstract
Bovine babesiosis caused by the protozoan parasite, Babesia bovis, remains a significant cause of avoidable economic losses to the livestock industry in many countries throughout the world. The molecular mechanisms underlying the pathophysiology of severe disease in susceptible cattle are not well understood and the tools available to study the biology of the parasite, including technologies for genetic manipulation, have only recently been developed. Recent availability of multiple parasite genomes and bioinformatic tools, in combination with the development of new biological reagents, will facilitate our better understanding of the parasite. This will ultimately assist in the identification of novel targets for the development of new therapeutics and vaccines. Here we describe some recent advances in Babesia research and highlight some important challenges for the future.
Collapse
Affiliation(s)
- Sejal Gohil
- Department of Microbiology, Monash University, Victoria 3800, Australia
| | | | | | | |
Collapse
|
30
|
|
31
|
Phylogenetic relationships of Mongolian Babesia bovis isolates based on the merozoite surface antigen (MSA)-1, MSA-2b, and MSA-2c genes. Vet Parasitol 2012; 184:309-16. [DOI: 10.1016/j.vetpar.2011.09.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 09/06/2011] [Accepted: 09/13/2011] [Indexed: 11/19/2022]
|
32
|
Genetic conservation of potentially immunogenic proteins among Brazilian isolates of Babesia bovis. Vet Parasitol 2012; 187:548-52. [PMID: 22309798 DOI: 10.1016/j.vetpar.2012.01.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Revised: 01/09/2012] [Accepted: 01/11/2012] [Indexed: 11/22/2022]
Abstract
Bovine babesiosis caused by Babesia bovis remains an important constraint for the development of cattle industries worldwide. Effective control can be achieved by vaccination with live attenuated phenotypes of the parasite. However, these vaccines have a number of drawbacks, which justifies the search for better, safer vaccines. In recent years, a number of parasite proteins with immunogenic potential have been discovered. However, there is little information on the genetic conservation of these proteins among different parasite isolates, which hinders their assessment as immunogens. The aim of the present study was to evaluate the conservation of the genes ama-1, acs-1, rap-1, trap, p0 and msa2c among five Brazilian isolates of B. bovis. Through polymerase chain reaction, genetic sequencing and bioinformatics analysis of the genes, a high degree of conservation (98-100%) was found among Brazilian isolates of B. bovis and the T2Bo isolate. Thus, these genes are worth considering as viable candidates to be included in a recombinant cocktail vaccine for cattle babesiosis caused by B. bovis.
Collapse
|
33
|
Suarez CE, Noh S. Emerging perspectives in the research of bovine babesiosis and anaplasmosis. Vet Parasitol 2011; 180:109-25. [DOI: 10.1016/j.vetpar.2011.05.032] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
34
|
Sun Y, Moreau E, Chauvin A, Malandrin L. The invasion process of bovine erythrocyte by Babesia divergens: knowledge from an in vitro assay. Vet Res 2011; 42:62. [PMID: 21569363 PMCID: PMC3117698 DOI: 10.1186/1297-9716-42-62] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Accepted: 05/11/2011] [Indexed: 11/10/2022] Open
Abstract
Babesia divergens is a tick-transmitted apicomplexan parasite for which asexual multiplication in its vertebrate hosts is restricted to erythrocytes. Current knowledge of invasion of these target cells is limited. An efficient in vitro invasion assay was set up to gain access to this information. Parasites prepared from infected RBC, lysed by electroporation, and mixed with bovine RBC in a selected synthetic medium (RPMI 1640 supplemented with calcium) were able to establish subsequent cultures with parasitemia ranging from 6 to 14%. Free parasites remaining in the invasion medium could be eliminated by Percoll gradient and culture could be pursued with the freshly invaded erythrocytes. In this way, the invasion time window could be shortened to obtain a synchronised start of the culture or to study the kinetics of invasion. With this assay we demonstrate that 1) erythrocyte invasion by B. divergens is a rapid process since 70% of the invasion-competent parasites invaded the RBC in less than 45 s; 2) all invasion-competent parasites achieved invasion within 10 min of contact; 3) one erythrocyte could be invaded concomitantly by two merozoites; 4) despite a synchronous start, the parasite population evolved heterogeneously resulting in a progressive loss of synchronisation. Western blot analysis of proteins collected from invasion medium were performed with sera from animals experimentally infected with B. divergens and highlighted several proteins. The dose-dependent, inhibitory effects of these sera on B. divergens invasion suggest that these proteins might be involved in the invasion process. Further investigations are required for their characterisation.
Collapse
Affiliation(s)
- Yi Sun
- ONIRIS, UMR1300, Biologie, Epidémiologie et Analyse de Risque en Santé Animale, Route de Gachet, La Chantrerie, BP 40706, F-44307 Nantes, France.
| | | | | | | |
Collapse
|
35
|
Gohil S, Kats LM, Sturm A, Cooke BM. Recent insights into alteration of red blood cells by Babesia bovis: moovin' forward. Trends Parasitol 2010; 26:591-9. [PMID: 20598944 DOI: 10.1016/j.pt.2010.06.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Revised: 06/07/2010] [Accepted: 06/08/2010] [Indexed: 10/19/2022]
Abstract
Over the past decade or so, our understanding of the biology of apicomplexan parasites has increased dramatically, particularly in the case of malaria. Notable achievements are the availability of complete genome sequences, transcriptome and proteome profiles and the establishment of in vitro transfection techniques for asexual-stage malaria parasites. Interestingly, despite their major economic importance and striking similarities with malaria, Babesia parasites have been relatively ignored, but change is on the horizon. Here, we bring together recent work on Babesia bovis parasites which are beginning to unravel the molecular mechanisms that underlie the pathogenesis of babesiosis and highlight some opportunities and challenges that lie ahead.
Collapse
Affiliation(s)
- Sejal Gohil
- Department of Microbiology, Monash University, Victoria, Australia
| | | | | | | |
Collapse
|
36
|
Using msa-2b as a molecular marker for genotyping Mexican isolates of Babesia bovis. INFECTION GENETICS AND EVOLUTION 2009; 9:1102-7. [DOI: 10.1016/j.meegid.2009.03.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2008] [Revised: 03/21/2009] [Accepted: 03/26/2009] [Indexed: 11/22/2022]
|
37
|
Dominguez M, Echaide I, Echaide STD, Mosqueda J, Cetrá B, Suarez CE, Florin-Christensen M. In silico predicted conserved B-cell epitopes in the merozoite surface antigen-2 family of B. bovis are neutralization sensitive. Vet Parasitol 2009; 167:216-26. [PMID: 19850413 DOI: 10.1016/j.vetpar.2009.09.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The merozoite surface antigens MSA-2 of Babesia bovis constitute a family of polymorphic GPI-anchored glycoproteins located at the parasite cell surface, that contain neutralization-sensitive B-cell epitopes. These are therefore putative vaccine candidates for bovine babesiosis. It was previously shown that (i) the MSA-2 antigens of the biologically cloned Mo7 strain are encoded by four tandemly organized genes: msa-2a(1), a(2), b and c, and (ii) at least one allele of each of these genes is present in the Argentine R1A strain with a moderate degree of polymorphism. The present work was aimed at defining neutralization-sensitive B-cell epitopes in the MSA-2 family, that are conserved among different B. bovis geographical isolates. To this end, msa-2a, b and c alleles from different isolates from Argentina, USA and Mexico were amplified by PCR, cloned and sequenced. Bioinformatic analysis by ClustalW alignments and B-cell epitope prediction algorithms performed on these sequences allowed the identification of several regions containing putative conserved B-cell epitopes. Four peptides representing these regions: (KDYKTMVKFCN from msa-2a(1); YYKKHIS, from msa-2b; and THDALKAVKQLIKT and ELLKLLIEA from msa-2c) were chemically synthesized, conjugated to keyhole limpet hemocyanin and used to inoculate mice to obtain immune sera. Anti-peptide antibodies recognized B. bovis merozoite extracts in all cases in ELISA tests. In addition, these sera reacted with the surface of merozoites of an Argentine and a Mexican B. bovis strains in immunofluorescence assays, and sera against two of the selected peptides inhibited invasion of erythrocytes by in vitro cultured merozoites. Taken together, the results show that the peptide sequences selected by bioinformatic analysis represent expressed and geographically conserved B. bovis B-cell epitopes that might be strong candidates for development of subunit vaccines.
Collapse
Affiliation(s)
- M Dominguez
- Institute of Pathobiology, Center of Agriculture and Veterinary Research, National Institute of Agriculture Technology, Castelar, Argentina
| | | | | | | | | | | | | |
Collapse
|
38
|
Chauvin A, Moreau E, Bonnet S, Plantard O, Malandrin L. Babesia and its hosts: adaptation to long-lasting interactions as a way to achieve efficient transmission. Vet Res 2009; 40:37. [PMID: 19379662 PMCID: PMC2695028 DOI: 10.1051/vetres/2009020] [Citation(s) in RCA: 161] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Accepted: 04/16/2009] [Indexed: 12/24/2022] Open
Abstract
Babesia, the causal agent of babesiosis, are tick-borne apicomplexan protozoa. True babesiae (Babesia genus sensu stricto) are biologically characterized by direct development in erythrocytes and by transovarial transmission in the tick. A large number of true Babesia species have been described in various vertebrate and tick hosts. This review presents the genus then discusses specific adaptations of Babesia spp. to their hosts to achieve efficient transmission. The main adaptations lead to long-lasting interactions which result in the induction of two reservoirs: in the vertebrate host during low long-term parasitemia and throughout the life cycle of the tick host as a result of transovarial and transstadial transmission. The molecular bases of these adaptations in vertebrate hosts are partially known but few of the tick-host interaction mechanisms have been elucidated.
Collapse
Affiliation(s)
- Alain Chauvin
- Ecole nationale vétérinaire, UMR 1300 BIOEPAR, ENVN, Atlanpôle - La Chantrerie, BP 40706, F-44307 Nantes Cedex 03, France.
| | | | | | | | | |
Collapse
|
39
|
Genis AD, Mosqueda JJ, Borgonio VM, Falcón A, Alvarez A, Camacho M, de Lourdes Muñoz M, Figueroa JV. Phylogenetic analysis of Mexican Babesia bovis isolates using msa and ssrRNA gene sequences. Ann N Y Acad Sci 2009; 1149:121-5. [PMID: 19120189 DOI: 10.1196/annals.1428.070] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Variable merozoite surface antigens of Babesia bovis are exposed glycoproteins having a role in erythrocyte invasion. Members of this gene family include msa-1 and msa-2 (msa-2c, msa-2a(1), msa-2a(2), and msa-2b). Small subunit ribosomal (ssr)RNA gene is subject to evolutive pressure and has been used in phylogenetic studies. To determine the phylogenetic relationship among B. bovis Mexican isolates using different genetic markers, PCR amplicons, corresponding to msa-1, msa-2c, msa-2b, and ssrRNA genes, were cloned and plasmids carrying the corresponding inserts were sequenced. Comparative analysis of nucleotide and deduced amino acid sequences revealed distinct degrees of variability and identity among the coding gene sequences obtained from 12 geographically different B. bovis isolates and a reference strain. Overall sequence identities of 47.7%, 72.3%, 87.7%, and 94% were determined for msa-1, msa-2b, msa-2c, and ssrRNA, respectively. A robust phylogenetic tree was obtained with msa-2b sequences. The phylogenetic analysis suggests that Mexican B. bovis isolates group in clades not concordant with the Mexican geography. However, the Mexican isolates group together in an American clade separated from the Australian clade. Sequence heterogeneity in msa-1, msa-2b, and msa-2c coding regions of Mexican B. bovis isolates present in different geographical regions can be a result of either differential evolutive pressure or cattle movement from commercial trade.
Collapse
Affiliation(s)
- Alma D Genis
- Posgrado en Ciencias Genomicas, Universidad Autonoma de la Ciudad de Mexico, México City, México
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Fish L, Leibovich B, Krigel Y, McElwain T, Shkap V. Vaccination of cattle against B. bovis infection with live attenuated parasites and non-viable immunogens. Vaccine 2008; 26 Suppl 6:G29-33. [DOI: 10.1016/j.vaccine.2008.09.070] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
41
|
Bork S, Yokoyama N, Hashiba S, Nakamura K, Takabatake N, Okamura M, Ikehara Y, Igarashi I. Asexual growth of Babesia bovis is inhibited by specific sulfated glycoconjugates. J Parasitol 2008; 93:1501-4. [PMID: 18314699 DOI: 10.1645/ge-1166.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In the present study, inhibitory effects of several sulfated and nonsulfated glycoconjugates were evaluated on the in vitro asexual growth of Babesia bovis. Among the selected sulfated glycoconjugates, dextran sulfate, heparin, heparan sulfate, fucoidan, and chondroitin sulfate B strongly inhibited the parasitic growth, and all but chondroitin sulfate B induced a significant accumulation of extracellular merozoites in culture. In contrast, chondroitin sulfate A, keratan sulfate, and protamine sulfate, as well as nonsulfated dextran and hyaluronic acid, did not influence the growth. These findings indicate that the asexual growth of B. bovis merozoites is inhibited by specific sulfated glycoconjugates, possibly providing us with an important insight into the molecular interaction(or interactions) during the process of the erythrocyte invasion by B. bovis merozoites.
Collapse
Affiliation(s)
- Sabine Bork
- Institute for Comparative Tropical Medicine and Parasitology, University of Munich, 80802 Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Howell JM, Ueti MW, Palmer GH, Scoles GA, Knowles DP. Transovarial transmission efficiency of Babesia bovis tick stages acquired by Rhipicephalus (Boophilus) microplus during acute infection. J Clin Microbiol 2006; 45:426-31. [PMID: 17166964 PMCID: PMC1829031 DOI: 10.1128/jcm.01757-06] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The protozoan parasite Babesia bovis, a reemerging threat to U.S. cattle, is acquired by adult female ticks of the subgenus Boophilus and is transovarially transmitted as the kinete stage to developing larval offspring. Sporozoites develop within larvae and are transmitted during larval feeding on a bovine host. This study evaluated the efficiency of B. bovis infection within Rhipicephalus (Boophilus) microplus following acquisition feeding on acutely parasitemic cattle. Parasite levels were quantified in blood from experimentally infected cattle and within hemolymph and larvae derived from acquisition-fed female B. microplus. There was a positive correlation between blood parasite levels in acutely parasitemic cattle and kinete levels in the hemolymph of adult female Boophilus ticks following acquisition feeding; however, there was no relationship between kinete levels in females and infection rates of larval progeny. Boophilus microplus females that acquisition fed produced larval progeny with infection rates of 12% to 48%. Importantly, larvae derived from replete females with very low levels of kinete infection, as demonstrated by microscopy and PCR, had infection rates of 22% to 30% and transmitted B. bovis during transmission feeding. These data demonstrate that although hemolymph infection may be undetectable, transmission to larval progeny occurs at a level which ensures transmission to the bovine host.
Collapse
Affiliation(s)
- Jeanne M Howell
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA 99164-7040, USA.
| | | | | | | | | |
Collapse
|
43
|
Brown WC, Norimine J, Goff WL, Suarez CE, McElwain TF. Prospects for recombinant vaccines against Babesia bovis and related parasites. Parasite Immunol 2006; 28:315-27. [PMID: 16842268 DOI: 10.1111/j.1365-3024.2006.00849.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Babesial parasites infect cattle in tropical and temperate regions of the world and cause significant morbidity and mortality. Discovery of protective antigens that could be used in a killed vaccine has been slow and to date there are few promising vaccine candidates for cattle Babesia. This review describes mechanisms of protective innate and adaptive immune responses to babesial parasites and different strategies to identify potentially protective protein antigens of B. bovis, B. bigemina, and B. divergens. Successful parasites often cause persistent infection, and this paper also discusses how B. bovis evades and regulates the immune response to promote survival of parasite and host. Development of successful non-living recombinant vaccines will depend on increased understanding of protective immune mechanisms and availability of parasite genomes.
Collapse
Affiliation(s)
- W C Brown
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA 99164-7040, USA.
| | | | | | | | | |
Collapse
|
44
|
LeRoith T, Berens SJ, Brayton KA, Hines SA, Brown WC, Norimine J, McElwain TF. The Babesia bovis merozoite surface antigen 1 hypervariable region induces surface-reactive antibodies that block merozoite invasion. Infect Immun 2006; 74:3663-7. [PMID: 16714599 PMCID: PMC1479293 DOI: 10.1128/iai.00032-06] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A hypervariable region (HVR) previously identified in the carboxy-terminal one-third of the Babesia bovis variable merozoite surface antigen family was more extensively analyzed in merozoite surface antigen 1 (MSA-1) from 16 strains and isolates. The MSA-1 HVR is proline rich and contains three semiconserved motifs nearly identical to those described for the related family member MSA-2. Two MSA-1-specific monoclonal antibodies previously shown to be reactive with the merozoite surface bound to a recombinant construct encoding the HVR, indicating that the HVR is surface exposed and accessible to antibody binding. Importantly, these surface-reactive, HVR-specific monoclonal antibodies were capable of inhibiting merozoite infectivity of the host erythrocyte in vivo. The results indicate that the MSA-1 HVR is involved in erythrocyte invasion and suggest that selection of MSA-1 variants may be driven by invasion-blocking antibodies.
Collapse
Affiliation(s)
- Tanya LeRoith
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington 99164-7040, USA.
| | | | | | | | | | | | | |
Collapse
|
45
|
Yokoyama N, Okamura M, Igarashi I. Erythrocyte invasion by Babesia parasites: Current advances in the elucidation of the molecular interactions between the protozoan ligands and host receptors in the invasion stage. Vet Parasitol 2006; 138:22-32. [PMID: 16504403 DOI: 10.1016/j.vetpar.2006.01.037] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
During an asexual growth cycle of Babesia parasites in a natural host, the extracellular merozoites invade (i.e., attach to, penetrate, and internalize) the host erythrocytes (RBC) via multiple adhesive interactions of several protozoan ligands with the target receptors on the host cell surface. After internalizing the host RBC, they asexually multiply, egress from the RBC by rupturing the host cells, and then invade the new RBC again. In the invasion stage, several surface-coating molecules of merozoites might be involved in the initial attachment to the RBC, while proteins secreted from apical organelles (rhoptry, microneme, and spherical body) are proposed to play roles mainly in erythrocyte penetration or internalization. On the other hand, several components located on the surface of the RBC, such as sialic acid residues, protease-sensitive proteins, or sulphated glycosaminoglycans, are identified or suspected as the host receptors of erythrocyte invasion by Babesia parasites. The detailed molecular interactions between Babesia merozoites and the host RBC are incompletely understood. In this review, these identified or suspected molecules (protozoan ligands/erythrocyte receptors) are described by especially focusing on Babesia bovis.
Collapse
Affiliation(s)
- Naoaki Yokoyama
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | | | | |
Collapse
|
46
|
Carcy B, Précigout E, Schetters T, Gorenflot A. Genetic basis for GPI-anchor merozoite surface antigen polymorphism of Babesia and resulting antigenic diversity. Vet Parasitol 2006; 138:33-49. [PMID: 16551492 DOI: 10.1016/j.vetpar.2006.01.038] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Glycosyl-phosphatidylinositol anchor merozoite surface antigens (GPI-anchor MSA) are proposed to act in the invasion process of infective merozoites of Babesia into host erythrocytes. Because of their essential function in the survival of Babesia parasites, they constitute good candidates for the development of vaccines against babesiosis and they have been extensively analyzed. These include Babesia bovis variable MSA (VMSA) and Babesia bigemina gp45/gp55 proteins of the agents of bovine babesiosis from tropical and subtropical countries, and the Babesia divergens Bd37 and Babesia canis Bc28 proteins of the main agents of bovine and canine babesiosis in Europe, respectively. However, these are very polymorphic antigens and Babesia parasites have evolved molecular mechanisms that enable these antigens to evade the host immune system as a survival strategy. This review focuses on the genetic basis of GPI-anchor MSA polymorphism and the antigenic diversity of B-cell epitopes that might be generated in each of these Babesia species. The picture is incomplete and no Babesia genome sequence is yet available. However, the available sequences suggest that two distinct, non cross-reactive GPI-anchor MSA (i.e., with unique B-cell epitopes) may be required by all Babesia species for invasion, and that these two distinct GPI-anchor MSA would be encoded by a multigene family. Furthermore, the data are consistent with the ability of biological clones from Babesia to use these multigene families for the expression of GPI-anchor MSA, either conserved (B. canis and B. bovis) or polymorphic (B. divergens and B. bigemina) in their amino acid sequence. Moreover, as a consequence for successful parasitism, the data suggest that both conserved and polymorphic GPI-anchor MSA would present unique B-cell epitopes.
Collapse
Affiliation(s)
- Bernard Carcy
- Laboratoire de Biologie Cellulaire et Moléculaire, EA MESR 2413, ERT 1038 Vaccination antiparasitaire, UFR des Sciences Pharmaceutiques et Biologiques, BP 14491, F-34093 Montpellier Cedex 5, France.
| | | | | | | |
Collapse
|
47
|
Abstract
Babesia bovis and its bovine host interact in many ways, resulting in a range of disease and infection phenotypes. Host responses to the parasite elicit or select for a variety of responses on the part of the parasite, the full range of which is not yet known. One well-established phenomenon, thought to aid parasite survival by evasion of host adaptive immune responses, is the sequential expansion of antigenically variant populations during an infection, a phenomenon referred to as "antigenic variation". Antigenic variation in B. bovis, like that in the human malarial parasite, Plasmodium falciparum, is intimately linked to a second survival mechanism, cytoadhesion. In cytoadhesion, mature parasite-containing erythrocytes bind to the capillary and post-capillary venous endothelium through parasite-derived ligands. The reliance of these parasites on both functions, and on their linkage, may provide opportunities to develop anti-babesial and, perhaps, anti-malarial protection strategies. The development of inhibitors of DNA metabolism in B. bovis may be used to abrogate the process of antigenic variation, whereas small molecular mimics may provide the means to vaccinate against a wide range of variants or to prevent the surface export of variant antigen ligands. In this article, aspects of antigenic variation and cytoadhesion in bovine babesiosis are explored, with a discussion of opportunities for prophylactic or therapeutic intervention in these intertwined processes.
Collapse
Affiliation(s)
- David R Allred
- Department of Pathobiology, University of Florida, Gainesville, FL 32611-0880, USA.
| | | |
Collapse
|
48
|
Norimine J, Ruef BJ, Palmer GH, Knowles DP, Herndon DR, Rice-Ficht AC, Brown WC. A novel 78-kDa fatty acyl-CoA synthetase (ACS1) of Babesia bovis stimulates memory CD4+ T lymphocyte responses in B. bovis-immune cattle. Mol Biochem Parasitol 2006; 147:20-9. [PMID: 16469396 DOI: 10.1016/j.molbiopara.2006.01.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2005] [Revised: 12/26/2005] [Accepted: 01/05/2006] [Indexed: 11/24/2022]
Abstract
Antigen-specific CD4+ T lymphocyte responses contribute to protective immunity against Babesia bovis, however the antigens that induce these responses remain largely unknown. A proteomic approach was used to identify novel B. bovis antigens recognized by memory CD4+ T cells from immune cattle. Fractions obtained from merozoites separated by continuous-flow electrophoresis (CFE) that contained proteins ranging from 20 to 83 kDa were previously shown to stimulate memory CD4+ lymphocyte responses in B. bovis-immune cattle. Expression library screening with rabbit antiserum raised against an immunostimulatory CFE fraction identified a clone encoding a predicted 78 kDa protein. BLAST analysis revealed sequence identity of this B. bovis protein with Plasmodium falciparum fatty acyl coenzyme A synthetase (ACS) family members (PfACS1-PfACS11), and the protein was designated B. bovis acyl-CoA synthetase 1 (ACS1). Southern blot analysis indicated that B. bovis ACS1 is encoded by a single gene, although BLAST analysis of the preliminary B. bovis genome sequence identified two additional family members, ACS2 and ACS3. Peripheral blood lymphocytes and CD4+ T cell lines from B. bovis-immune cattle proliferated significantly against recombinant ACS1 protein, consistent with its predicted involvement in protective immunity. However, immune sera from cattle recovered from B. bovis infection did not react with ACS1, indicating that epitopes may be conformationally dependent.
Collapse
Affiliation(s)
- Junzo Norimine
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA 99164-7040, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Berens SJ, Brayton KA, Molloy JB, Bock RE, Lew AE, McElwain TF. Merozoite surface antigen 2 proteins of Babesia bovis vaccine breakthrough isolates contain a unique hypervariable region composed of degenerate repeats. Infect Immun 2005; 73:7180-9. [PMID: 16239512 PMCID: PMC1273907 DOI: 10.1128/iai.73.11.7180-7189.2005] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The merozoite surface antigen 2 (MSA-2) proteins of Babesia bovis are members of the variable merozoite surface antigen (VMSA) family that have been implicated in erythrocyte invasion and are important targets for antibody-mediated blocking of invasion. Extensive sequence variation in another VMSA member, MSA-1, has been shown in all vaccine breakthrough isolates. To test the hypothesis that the msa-2 genes of vaccine breakthrough isolates would also encode a diverse set of proteins, the complete msa-2 locus was characterized from 12 Australian B. bovis strains and isolates, including two vaccine strains and eight vaccine breakthrough isolates, and compared to the loci in previously and newly characterized American strains. In contrast to American strains, the msa-2 loci of all Australian strains and isolates examined contain, in addition to msa-2c, only a solitary gene (designated msa-2a/b) closely related to American strain msa-2a and msa-2b. Nevertheless, the proteins encoded by these genes are quite diverse both between and within geographic regions and harbor evidence of genetic exchange among other VMSA family members, including msa-1. Moreover, all but one of the Australian breakthrough isolate MSA-2a/b proteins is markedly different from the vaccine strain from which immune escape occurred, consistent with their role in strain-specific protective immunity. The densest distribution of polymorphisms occurs in a hypervariable region (HVR) within the carboxy third of the molecule that is highly proline rich. Variation in length and content of the HVR is primarily attributable to differences in the order and number of degenerate nucleotide repeats encoding three motifs of unknown function.
Collapse
Affiliation(s)
- Shawn J Berens
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA 99164-7040, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Cooke BM, Mohandas N, Cowman AF, Coppel RL. Cellular adhesive phenomena in apicomplexan parasites of red blood cells. Vet Parasitol 2005; 132:273-95. [PMID: 16087297 DOI: 10.1016/j.vetpar.2005.07.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The apicomplexan parasites Babesia and Plasmodium are related, yet phylogenetically distinct haemoprotozoa that infect red blood cells and cause severe diseases of major human and veterinary importance. A variety of cellular and molecular interactions are pivotal in many aspects of the pathogenicity of these two parasites. Comparison of the cellular and molecular mechanisms that culminate in accumulation of parasitised red blood cells in the microvasculature of cattle infected with Babesia bovis (babesiosis) and humans infected with Plasmodium falciparum (falciparum malaria) is particularly instructive given the striking similarities in the pathophysiology of these two important medical and veterinary diseases. While such adhesive phenomena have been studied extensively in malaria, they have received relatively little attention in babesiosis. In this review, we summarise the findings of more than 25 years of research into cellular adhesive phenomena in malaria and speculate on how this body of work can now be applied to Babesia parasites. Such information is fundamental if we are to learn more about the biology of Babesia parasites, the cellular and molecular mechanisms by which they cause infection and disease and how to develop novel therapeutic strategies or vaccines for both Babesia and malaria infections.
Collapse
Affiliation(s)
- Brian M Cooke
- Department of Microbiology, Monash University, Vic. 3800, Australia.
| | | | | | | |
Collapse
|