1
|
Li DF, Wang S, Suarez CE, Xuan X, He L, Zhao JL. Pushing the frontiers of babesiosis research: in vitro culture and gene editing. Trends Parasitol 2025; 41:317-329. [PMID: 40089452 DOI: 10.1016/j.pt.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 03/17/2025]
Abstract
Babesiosis is a tick-borne parasitic disease that poses a significant risk to both animal and human health. A comprehensive understanding of Babesia biology necessitates the application of advanced laboratory techniques. This review explores recent advancements in gene editing technologies of Babesia, emphasizing the foundational importance of in vitro culture systems. We highlight the historical challenges encountered in establishing effective in vitro culture and discuss the need for optimizing these methods to enhance gene editing efficiency. Here, we describe recent progress in Babesia transfection, different gene manipulation systems, and the applications of gene editing. This review aims to provide essential insights and technical guidance for future studies in Babesia genetics, highlighting the transformative potential of gene manipulation in combating this important parasitic disease.
Collapse
Affiliation(s)
- Dong-Fang Li
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Wuhan, Hubei Province 430070, PR China
| | - Sen Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Wuhan, Hubei Province 430070, PR China
| | - Carlos E Suarez
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Xuenan Xuan
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan; Research Center for Asian Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
| | - Lan He
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province 430070, PR China; Key Laboratory of Animal Epidemical Disease and Infectious Zoonoses, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei Province 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Wuhan, Hubei Province 430070, PR China.
| | - Jun-Long Zhao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province 430070, PR China; Key Laboratory of Animal Epidemical Disease and Infectious Zoonoses, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei Province 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Wuhan, Hubei Province 430070, PR China.
| |
Collapse
|
2
|
Webi E, Abkallo HM, Obiero G, Ndegwa P, Xie S, Zhao S, Nene V, Steinaa L. Genome Editing in Apicomplexan Parasites: Current Status, Challenges, and Future Possibilities. CRISPR J 2024; 7:310-326. [PMID: 39387255 DOI: 10.1089/crispr.2024.0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024] Open
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein (Cas) technology has revolutionized genome editing across various biological systems, including the Apicomplexa phylum. This review describes the status, challenges, and applications of CRISPR-Cas9 editing technology in apicomplexan parasites, such as Plasmodium, Toxoplasma, Theileria, Babesia, and Cryptosporidium. The discussion encompasses successfully implemented CRISPR-Cas9-based techniques in these parasites, highlighting the achieved milestones, from precise gene modifications to genome-wide screening. In addition, the review addresses the challenges hampering efficient genome editing, including the parasites' complex life cycles, multiple intracellular stages, and the lack of robust genetic tools. It further explores the ethical and policy considerations surrounding genome editing and the future perspectives of CRISPR-Cas applications in apicomplexan parasites.
Collapse
Affiliation(s)
- Ethel Webi
- Animal and Human Health Program, International Livestock Research Institute, Nairobi, Kenya
- Department of Biochemistry, University of Nairobi, Nairobi, Kenya
| | - Hussein M Abkallo
- Animal and Human Health Program, International Livestock Research Institute, Nairobi, Kenya
| | - George Obiero
- Department of Biochemistry, University of Nairobi, Nairobi, Kenya
| | - Paul Ndegwa
- Department of Biology, University of Nairobi, Nairobi, Kenya
| | - Shengsong Xie
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, P. R. China
| | - Shuhong Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, P. R. China
| | - Vishvanath Nene
- Animal and Human Health Program, International Livestock Research Institute, Nairobi, Kenya
| | - Lucilla Steinaa
- Animal and Human Health Program, International Livestock Research Institute, Nairobi, Kenya
| |
Collapse
|
3
|
ARAYASKUL N, ASADA M, FATHI A, ARIEFTA NR, KOMATSU K, SUGANUMA K, INOUE N, KAWAZU SI. Stable expression of red fluorescent protein-blasticidin deaminase fusion gene (rfp-bsd) as a selectable marker for DNA transfection in Babesia ovata. J Vet Med Sci 2024; 86:744-747. [PMID: 38749739 PMCID: PMC11251815 DOI: 10.1292/jvms.24-0111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 04/28/2024] [Indexed: 07/05/2024] Open
Abstract
The red fluorescent protein (rfp)-blasticidin deaminase (bsd) fusion gene was transfected into Babesia ovata by electroporation with the plasmid DNA and selected with 15 μg/mL of blasticidin S under the in vitro culture condition. The transfected parasite with episomal DNA was selected and cultured for further analysis based on the presence of the rfp-bsd fusion gene by PCR and expression of the fusion protein by immunofluorescence antibody test under fluorescence microscopy for 2 months after the transfection. The results are the first, to our knowledge, to demonstrate the expression and stability of the episomal rfp-bsd fusion gene under the control of actin promoter as a selectable marker for the transfection system in B. ovata.
Collapse
Affiliation(s)
- Nada ARAYASKUL
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
| | - Masahito ASADA
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
| | - Atefeh FATHI
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
| | - Nanang R ARIEFTA
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
| | - Kota KOMATSU
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
| | - Keisuke SUGANUMA
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
| | - Noboru INOUE
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
| | - Shin-ichiro KAWAZU
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Japan
| |
Collapse
|
4
|
Rojas MJ, Bastos RG, Navas J, Laughery JM, Lacy PA, Suarez CE. A conserved motif in the immune-subdominant RAP-1 related antigen of Babesia bovis contains a B-cell epitope recognized by antibodies from protected cattle. Front Immunol 2024; 15:1380660. [PMID: 38720894 PMCID: PMC11076753 DOI: 10.3389/fimmu.2024.1380660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/10/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction Babesia bovis, a tick-borne apicomplexan parasite causing bovine babesiosis, remains a significant threat worldwide, and improved and practical vaccines are needed. Previous studies defined the members of the rhoptry associated protein-1 (RAP-1), and the neutralization-sensitive rhoptry associated protein-1 related antigen (RRA) superfamily in B. bovis, as strong candidates for the development of subunit vaccines. Both RAP-1 and RRA share conservation of a group of 4 cysteines and amino acids motifs at the amino terminal end (NT) of these proteins. Methods and results Sequence comparisons among the RRA sequences of several B. bovis strains and other Babesia spp parasites indicate a high level of conservation of a 15-amino acid (15-mer) motif located at the NT of the protein. BlastP searches indicate that the 15-mer motif is also present in adenylate cyclase, dynein, and other ATP binding proteins. AlphaFold2 structure predictions suggest partial exposure of the 15-mer on the surface of RRA of three distinct Babesia species. Antibodies in protected cattle recognize a synthetic peptide representing the 15-mer motif sequence in iELISA, and rabbit antibodies against the 15-mer react with the surface of free merozoites in immunofluorescence. Discussion and conclusion The presence of the 15-mer-like regions in dynein and ATP-binding proteins provides a rationale for investigating possible functional roles for RRA. The demonstrated presence of a surface exposed B-cell epitope in the 15-mer motif of the B. bovis RRA, which is recognized by sera from protected bovines, supports its inclusion in future subunit epitope-based vaccines against B. bovis.
Collapse
Affiliation(s)
- Manuel J. Rojas
- Department of Veterinary Microbiology & Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
- Animal Health Department, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Reginaldo G. Bastos
- Department of Veterinary Microbiology & Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
- Animal Disease Research Unit, Agricultural Research Service, United States Department of Agriculture, Pullman, WA, United States
| | - Jinna Navas
- Department of Veterinary Microbiology & Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Jacob M. Laughery
- Department of Veterinary Microbiology & Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Paul A. Lacy
- Animal Disease Research Unit, Agricultural Research Service, United States Department of Agriculture, Pullman, WA, United States
| | - Carlos E. Suarez
- Department of Veterinary Microbiology & Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
- Animal Disease Research Unit, Agricultural Research Service, United States Department of Agriculture, Pullman, WA, United States
| |
Collapse
|
5
|
Groomes PV, Paul AS, Duraisingh MT. Inhibition of malaria and babesiosis parasites by putative red blood cell targeting small molecules. Front Cell Infect Microbiol 2024; 14:1304839. [PMID: 38572319 PMCID: PMC10988762 DOI: 10.3389/fcimb.2024.1304839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 02/15/2024] [Indexed: 04/05/2024] Open
Abstract
Background Chemotherapies for malaria and babesiosis frequently succumb to the emergence of pathogen-related drug-resistance. Host-targeted therapies are thought to be less susceptible to resistance but are seldom considered for treatment of these diseases. Methods Our overall objective was to systematically assess small molecules for host cell-targeting activity to restrict proliferation of intracellular parasites. We carried out a literature survey to identify small molecules annotated for host factors implicated in Plasmodium falciparum infection. Alongside P. falciparum, we implemented in vitro parasite susceptibility assays also in the zoonotic parasite Plasmodium knowlesi and the veterinary parasite Babesia divergens. We additionally carried out assays to test directly for action on RBCs apart from the parasites. To distinguish specific host-targeting antiparasitic activity from erythrotoxicity, we measured phosphatidylserine exposure and hemolysis stimulated by small molecules in uninfected RBCs. Results We identified diverse RBC target-annotated inhibitors with Plasmodium-specific, Babesia-specific, and broad-spectrum antiparasitic activity. The anticancer MEK-targeting drug trametinib is shown here to act with submicromolar activity to block proliferation of Plasmodium spp. in RBCs. Some inhibitors exhibit antimalarial activity with transient exposure to RBCs prior to infection with parasites, providing evidence for host-targeting activity distinct from direct inhibition of the parasite. Conclusions We report here characterization of small molecules for antiproliferative and host cell-targeting activity for malaria and babesiosis parasites. This resource is relevant for assessment of physiological RBC-parasite interactions and may inform drug development and repurposing efforts.
Collapse
Affiliation(s)
| | | | - Manoj T. Duraisingh
- Department of Immunology & Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, United States
| |
Collapse
|
6
|
Sitaraman R. Subversion from Within and Without: Effector Molecule Transfer from Obligate Intracellular Apicomplexan Parasites to Human Host Cells. Results Probl Cell Differ 2024; 73:521-535. [PMID: 39242391 DOI: 10.1007/978-3-031-62036-2_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2024]
Abstract
Intracellular protozoan pathogens have to negotiate the internal environment of the host cell they find themselves in, as well as manipulate the host cell to ensure their own survival, replication, and dissemination. The transfer of key effector molecules from the pathogen to the host cell is crucial to this interaction and is technically more demanding to study as compared to an extracellular pathogen. While several effector molecules have been identified, the mechanisms and conditions underlying their transfer to the host cell remain partly or entirely unknown. Improvements in experimental systems have revealed tantalizing details of such intercellular transfer, which form the subject of this chapter.
Collapse
|
7
|
Hötzel I, Suarez CE. Structural definition of babesial RAP-1 proteins identifies a novel protein superfamily across Apicomplexa. Sci Rep 2023; 13:22330. [PMID: 38102310 PMCID: PMC10724250 DOI: 10.1038/s41598-023-49532-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023] Open
Abstract
Apicomplexan protozoa are intracellular parasites of medical and economic importance. These parasites contain specialized apical complex organelles, including rhoptries, that participate in the process of host cell invasion. Conserved antigens expressed in the rhoptries are rational vaccine targets, but whether conservation of protein structure is a functional requirement for invasion remains unknown. Novel protein structural modeling enables identification of structurally conserved protein families that are not evident by sequence analysis alone. Here we show by AlphaFold2 structural modeling that the rhoptry-associated protein 1 superfamily of the Piroplasmida hemoparasites Babesia and Theileria (pRAP-1) is structurally conserved, with the core conserved region being composed of a globin-like and a 4-helix bundle subdomain. Search for structurally related members of this protein family in other apicomplexan parasites revealed structural homologues of pRAP-1 in several species of Plasmodium, Toxoplasma gondii and other members of the Sarcocystidae family. Based on these structural findings, pRAP-1 is a conserved apical complex protein, but whether these proteins share functional features in different species remains unknown. Identification of widely conserved elements involved in infection in these parasites will enhance our knowledge of invasion mechanisms, and facilitate the design of methods for controlling diseases that affect humans and animals globally.
Collapse
Affiliation(s)
- Isidro Hötzel
- Department of Antibody Engineering, Genentech, South San Francisco, CA, 94080, USA
| | - Carlos E Suarez
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA.
- Animal Disease Research Unit, United States Department of Agriculture - Agricultural Research Service, Pullman, WA, USA.
| |
Collapse
|
8
|
Cubillos EFG, Snebergerova P, Borsodi S, Reichensdorferova D, Levytska V, Asada M, Sojka D, Jalovecka M. Establishment of a stable transfection and gene targeting system in Babesia divergens. Front Cell Infect Microbiol 2023; 13:1278041. [PMID: 38156314 PMCID: PMC10753763 DOI: 10.3389/fcimb.2023.1278041] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/29/2023] [Indexed: 12/30/2023] Open
Abstract
Babesia divergens is an emerging tick-borne pathogen considered as the principal causative agent of bovine babesiosis in Europe with a notable zoonotic risk to human health. Despite its increasing impact, considerable gaps persist in our understanding of the molecular interactions between this parasite and its hosts. In this study, we address the current limitation of functional genomic tools in B. divergens and introduce a stable transfection system specific to this parasite. We define the parameters for a drug selection system hdhfr-WR99210 and evaluate different transfection protocols for highly efficient generation of transgenic parasites expressing GFP. We proved that plasmid delivery into bovine erythrocytes prior to their infection is the most optimal transfection approach for B. divergens, providing novel evidence of Babesia parasites' ability to spontaneously uptake external DNA from erythrocytes cytoplasm. Furthermore, we validated the bidirectional and symmetrical activity of ef-tgtp promoter, enabling simultaneous expression of external genes. Lastly, we generated a B. divergens knockout line by targeting a 6-cys-e gene locus. The observed dispensability of this gene in intraerythrocytic parasite development makes it a suitable recipient locus for further transgenic application. The platform for genetic manipulations presented herein serves as the initial step towards developing advanced functional genomic tools enabling the discovery of B. divergens molecules involved in host-vector-pathogen interactions.
Collapse
Affiliation(s)
- Eliana F. G. Cubillos
- Faculty of Science, University of South Bohemia in Ceske Budejovice, Ceske Budejovice, Czechia
| | - Pavla Snebergerova
- Faculty of Science, University of South Bohemia in Ceske Budejovice, Ceske Budejovice, Czechia
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czechia
| | - Sarka Borsodi
- Faculty of Science, University of South Bohemia in Ceske Budejovice, Ceske Budejovice, Czechia
| | | | - Viktoriya Levytska
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czechia
| | - Masahito Asada
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Hokkaido, Obihiro, Japan
| | - Daniel Sojka
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czechia
| | - Marie Jalovecka
- Faculty of Science, University of South Bohemia in Ceske Budejovice, Ceske Budejovice, Czechia
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czechia
| |
Collapse
|
9
|
Andrews M, Baum J, Gilson PR, Wilson DW. Bottoms up! Malaria parasite invasion the right way around. Trends Parasitol 2023; 39:1004-1013. [PMID: 37827961 DOI: 10.1016/j.pt.2023.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 10/14/2023]
Abstract
A critical part of the malaria parasite's life cycle is invasion of red blood cells (RBCs) by merozoites. Inside RBCs, the parasite forms a schizont, which undergoes segmentation to produce daughter merozoites. These cells are released, establishing cycles of invasion. Traditionally, merozoites are represented as nonmotile, egg-shaped cells that invade RBCs 'narrower end' first and pack within schizonts with this narrower end facing outwards. Here, we discuss recent evidence and re-evaluate previous data which suggest that merozoites are capable of motility and have spherical or elongated-teardrop shapes. Furthermore, merozoites invade RBCs 'wider end' first and pack within schizonts with this wider end facing outwards. We encourage the field to review this revised model and consider its implications for future studies.
Collapse
Affiliation(s)
- Mia Andrews
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide 5005, Australia
| | - Jake Baum
- School of Biomedical Sciences, UNSW Sydney, Kensington, NSW 2052, Australia; Department of Life Sciences, Imperial College London, South Kensington, London SW7 2AZ, UK
| | - Paul R Gilson
- Burnet Institute, Melbourne 3004, Victoria, Australia; Department of Microbiology and Immunology, The University of Melbourne, Melbourne 3010, Victoria, Australia
| | - Danny W Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide 5005, Australia; Burnet Institute, Melbourne 3004, Victoria, Australia; Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, 5005, SA, Australia.
| |
Collapse
|
10
|
Kuleš J, Rubić I, Farkaš V, Barić Rafaj R, Gotić J, Crnogaj M, Burchmore R, Eckersall D, Mrljak V, Leisewitz AL. Serum proteome profiling of naturally acquired Babesia rossi infection in dogs. Sci Rep 2023; 13:10249. [PMID: 37353646 PMCID: PMC10290096 DOI: 10.1038/s41598-023-37312-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023] Open
Abstract
Babesiosis is a disease of significant medically and veterinary importance with worldwide distribution. It is caused by intra-erythrocyte protozoal parasites, with Babesia rossi causing the most severe clinical signs of all the large Babesia parasites infecting dogs. The disease can be clinically classified into uncomplicated and complicated forms with a wide range of clinical presentations from a mild, subclinical illness to complicated forms and death. The aim of this study was to assess serum proteomic profiles from dogs with babesiosis and healthy dogs using a label-based proteomics approach. Altogether 32 dogs naturally infected with B. rossi (subdivided into 18 uncomplicated cases and 14 complicated cases of babesiosis) and 20 healthy dogs were included. There were 78 proteins with significantly different abundances between the three groups of dogs. Elucidation of proteins and pathways involved in canine babesiosis caused by B. rossi have revealed key differences associated with haemostasis, innate immune system, lipid metabolism and inflammation. Shotgun proteomic profiling allowed identification of potential serum biomarkers for differentiation of disease severity in canine babesiosis caused by B. rossi. These findings may be applicable to the study of host-parasite interactions and the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Josipa Kuleš
- Department of Chemistry and Biochemistry, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Ivana Rubić
- Laboratory of Proteomics, Faculty of Veterinary Medicine, Internal Diseases Clinic, University of Zagreb, Zagreb, Croatia
| | - Vladimir Farkaš
- Laboratory of Proteomics, Faculty of Veterinary Medicine, Internal Diseases Clinic, University of Zagreb, Zagreb, Croatia
| | - Renata Barić Rafaj
- Department of Chemistry and Biochemistry, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Jelena Gotić
- Laboratory of Proteomics, Faculty of Veterinary Medicine, Internal Diseases Clinic, University of Zagreb, Zagreb, Croatia
| | - Martina Crnogaj
- Laboratory of Proteomics, Faculty of Veterinary Medicine, Internal Diseases Clinic, University of Zagreb, Zagreb, Croatia
| | - Richard Burchmore
- Glasgow Polyomics, College of Veterinary, Medical and Life Sciences, University of Glasgow, Glasgow, UK
| | - David Eckersall
- College of Veterinary, Medical and Life Sciences, School of Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Vladimir Mrljak
- Laboratory of Proteomics, Faculty of Veterinary Medicine, Internal Diseases Clinic, University of Zagreb, Zagreb, Croatia.
| | - Andrew L Leisewitz
- Department of Clinical Sciences, Auburn University College of Veterinary Medicine, Auburn, AL, USA
- Department of Companion Animal Clinical Studies, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, South Africa
| |
Collapse
|
11
|
Oladokun R, Adekanmbi E, Ueti M, Srivastava S. Dielectric characterization of Babesia bovis using the dielectrophoretic crossover frequency. Electrophoresis 2023. [PMID: 37160713 DOI: 10.1002/elps.202200263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 04/12/2023] [Accepted: 04/19/2023] [Indexed: 05/11/2023]
Abstract
Coinfection with the tick-transmitted pathogen Babesia spp. is becoming a serious health problem because of the erythrocyte invasion through Ixodes scapularis tick. The transmission of this protozoan by blood transfusion often results in high morbidity and mortality in recipients. A novel way to detect parasitized erythrocytes is by utilizing dielectrophoresis, an electrokinetic technique on a microfluidic platform, to improve the diagnostics of Babesia spp. The differences in the dielectric properties of Babesia spp.-infected erythrocytes versus healthy erythrocytes were exploited to design a fast and cost-effective diagnostic tool. One crucial factor for a successful diagnostic platform via dielectrophoretic separation is the dielectric characterization of Babesia-infected erythrocytes, which is investigated in this paper. The influence of medium conductivity and erythrocytes phenotype and genotype over the first crossover frequency (fco1 ) are used to quantify the dielectric properties of the infected cells. A sigmoidal curve was plotted via curve fitting of the single-shell model, which has been proven appropriate for parasitized cell populations where considerable cell geometry variation occurs. The difference in these curves is relevant for the separation of cells population. Microliters of sample and reagent were used throughout this experiment; the scale, results obtained, and simplicity of the system often make it very suitable for point-of-care babesiosis disease diagnostics.
Collapse
Affiliation(s)
- Raphael Oladokun
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, West Virginia, USA
| | | | - Massaro Ueti
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Soumya Srivastava
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
12
|
Wang J, Chen K, Ren Q, Zhang S, Yang J, Wang Y, Nian Y, Li X, Liu G, Luo J, Yin H, Guan G. Comparative genomics reveals unique features of two Babesia motasi subspecies: Babesia motasi lintanensis and Babesia motasi hebeiensis. Int J Parasitol 2023; 53:265-283. [PMID: 37004737 DOI: 10.1016/j.ijpara.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 02/05/2023] [Accepted: 02/12/2023] [Indexed: 04/03/2023]
Abstract
Parasites of the Babesia genus are prevalent worldwide and infect a wide diversity of domestic animals and humans. Herein, using Oxford Nanopore Technology and Illumina sequencing technologies, we sequenced two Babesia sub-species, Babesia motasi lintanensis and Babesia motasi hebeiensis. We identified 3,815 one-to-one ortholog genes that are specific to ovine Babesia spp. Phylogenetic analysis reveals that the two B. motasi subspecies form a distinct clade from other Piroplasma spp. Consistent with their phylogenetic position, comparative genomic analysis reveals that these two ovine Babesia spp. share higher colinearity with Babesia bovis than with Babesia microti. Concerning the speciation date, B. m. lintanensis split from B. m. hebeiensis approximately 17 million years ago. Genes correlated to transcription, translation, protein modification and degradation, as well as differential/specialized gene family expansions in these two subspecies may favor adaptation to vertebrate and tick hosts. The close relationship between B. m. lintanensis and B. m. hebeiensis is underlined by a high degree of genomic synteny. Compositions of most invasion, virulence, development, and gene transcript regulation-related multigene families, including spherical body protein, variant erythrocyte surface antigen, glycosylphosphatidylinositol anchored proteins, and transcription factor Apetala 2 genes, is largely conserved, but in contrast to this conserved situation, we observe major differences in species-specific genes that may be involved in multiple functions in parasite biology. For the first time in Babesia spp., we find abundant fragments of long terminal repeat-retrotransposons in these two species. We provide fundamental information to characterize the genomes of B. m. lintanensis and B. m. hebeiensis, providing insights into the evolution of B. motasi group parasites.
Collapse
Affiliation(s)
- Jinming Wang
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, Gansu 730046, China.
| | - Kai Chen
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| | - Qiaoyun Ren
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, Gansu 730046, China.
| | - Shangdi Zhang
- Department of Clinical Laboratory, The Second Hospital of Lanzhou University, Lanzhou, China.
| | - Jifei Yang
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, Gansu 730046, China.
| | - Yanbo Wang
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, Gansu 730046, China; Department of Clinical Laboratory, The Second Hospital of Lanzhou University, Lanzhou, China.
| | - Yueli Nian
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, Gansu 730046, China; Department of Clinical Laboratory, The Second Hospital of Lanzhou University, Lanzhou, China.
| | - Xiaoyun Li
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, Gansu 730046, China.
| | - Guangyuan Liu
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, Gansu 730046, China.
| | - Jianxun Luo
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, Gansu 730046, China.
| | - Hong Yin
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, Gansu 730046, China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou 225009, China.
| | - Guiquan Guan
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, Gansu 730046, China.
| |
Collapse
|
13
|
Song P, Cai YC, Chen MX, Chen SH, Chen JX. Enhanced phosphatidylserine exposure and erythropoiesis in Babesia microti-infected mice. Front Microbiol 2023; 13:1083467. [PMID: 36687590 PMCID: PMC9846230 DOI: 10.3389/fmicb.2022.1083467] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/08/2022] [Indexed: 12/02/2023] Open
Abstract
INTRODUCTION Babesia microti (B. microti) is the dominant species responsible for human babesiosis, which is associated with severe hemolytic anemia and splenomegaly because it infects mammalian erythrocytes. The actual prevalence of B. microti is thought to have been substantially underestimated. METHODS In this study, Bagg's albino/c (BALB/c) mice were intraperitoneally injected with B. microti-infected erythrocytes, and parasitemia was subsequently measured by calculating the proportion of infected erythrocytes. The ultrastructure of infected erythrocytes was observed using scanning and transmission electron microscopes. Quantifying phosphatidylserine (PS) exposure, oxidative stress, intracellular Ca2+, and erythropoiesis of erythrocytes were done using flow cytometry. The physiological indicators were analyzed using a Mindray BC-5000 Vet automatic hematology analyzer. RESULTS Of note, 40.7 ± 5.9% of erythrocytes changed their structure and shrunk in the B. microti-infected group. The percentage of annexin V-positive erythrocytes and the levels of reactive oxygen species (ROS) in the erythrocytes were higher in the B. microti-infected group than in the control group at 10 dpi. Significant splenomegaly and severe anemia were also observed following B. microti infection. The parasitemia level in the B. microti-infected splenectomized group was higher than that of the B. microti-infected sham group. The population of early erythroblasts increased, and the late erythroblasts decreased in both the bone marrow and spleen tissues of the B. microti-infected group at 10 dpi. DISCUSSION PS exposure and elevated ROS activities were hallmarks of eryptosis in the B. microti-infected group. This study revealed for the first time that B. microti could also induce eryptosis. At the higher parasitemia phase, the occurrence of severe anemia and significant changes in the abundance of erythroblasts in B. microti-infected mice group were established. The spleen plays a critical protective role in controlling B. microti infection and preventing anemia. B. microti infection could cause a massive loss of late erythroblasts and induce erythropoiesis.
Collapse
Affiliation(s)
- Peng Song
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
- NHC Key Laboratory of Parasite and Vector Biology, Ministry of Public Health, Shanghai, China
- WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China
- Hainan Tropical Diseases Research Center (Chinese Center for Tropical Diseases Research, Hainan), Haikou, Hainan, China
| | - Yu-Chun Cai
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
- NHC Key Laboratory of Parasite and Vector Biology, Ministry of Public Health, Shanghai, China
- WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China
| | - Mu-Xin Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
- NHC Key Laboratory of Parasite and Vector Biology, Ministry of Public Health, Shanghai, China
- WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China
- Hainan Tropical Diseases Research Center (Chinese Center for Tropical Diseases Research, Hainan), Haikou, Hainan, China
| | - Shao-Hong Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
- NHC Key Laboratory of Parasite and Vector Biology, Ministry of Public Health, Shanghai, China
- WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China
| | - Jia-Xu Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
- NHC Key Laboratory of Parasite and Vector Biology, Ministry of Public Health, Shanghai, China
- WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China
- Hainan Tropical Diseases Research Center (Chinese Center for Tropical Diseases Research, Hainan), Haikou, Hainan, China
| |
Collapse
|
14
|
Capelli-Peixoto J, Saelao P, Johnson WC, Kappmeyer L, Reif KE, Masterson HE, Taus NS, Suarez CE, Brayton KA, Ueti MW. Comparison of high throughput RNA sequences between Babesia bigemina and Babesia bovis revealed consistent differential gene expression that is required for the Babesia life cycle in the vertebrate and invertebrate hosts. Front Cell Infect Microbiol 2022; 12:1093338. [PMID: 36601308 PMCID: PMC9806345 DOI: 10.3389/fcimb.2022.1093338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Bovine babesiosis caused by Babesia bigemina and Babesia bovis is an economically important disease that affects cattle worldwide. Both B. bigemina and B. bovis are transovarially transmitted by Rhipicephalus ticks. However, little is known regarding parasite gene expression during infection of the tick vector or mammalian host, which has limited the development of effective control strategies to alleviate the losses to the cattle industry. To understand Babesia gene regulation during tick and mammalian host infection, we performed high throughput RNA-sequencing using samples collected from calves and Rhipicephalus microplus ticks infected with B. bigemina. We evaluated gene expression between B. bigemina blood-stages and kinetes and compared them with previous B. bovis RNA-seq data. The results revealed similar patterns of gene regulation between these two tick-borne transovarially transmitted Babesia parasites. Like B. bovis, the transcription of several B. bigemina genes in kinetes exceeded a 1,000-fold change while a few of these genes had a >20,000-fold increase. To identify genes that may have important roles in B. bigemina and B. bovis transovarial transmission, we searched for genes upregulated in B. bigemina kinetes in the genomic datasets of B. bovis and non-transovarially transmitted parasites, Theileria spp. and Babesia microti. Using this approach, we identify genes that may be potential markers for transovarial transmission by B. bigemina and B. bovis. The findings presented herein demonstrate common Babesia genes linked to infection of the vector or mammalian host and may contribute to elucidating strategies used by the parasite to complete their life cycle.
Collapse
Affiliation(s)
- Janaina Capelli-Peixoto
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States,*Correspondence: Janaina Capelli-Peixoto,
| | - Perot Saelao
- Veterinary Pest Genetic Research Unit, USDA-ARS, Kerrville, TX, United States
| | | | - Lowell Kappmeyer
- Animal Disease Research Unit, USDA-ARS, Pullman, WA, United States
| | - Kathryn E. Reif
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Hayley E. Masterson
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Naomi S. Taus
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States,Animal Disease Research Unit, USDA-ARS, Pullman, WA, United States
| | - Carlos E. Suarez
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States,Animal Disease Research Unit, USDA-ARS, Pullman, WA, United States
| | - Kelly A. Brayton
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Massaro W. Ueti
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, United States,Animal Disease Research Unit, USDA-ARS, Pullman, WA, United States
| |
Collapse
|
15
|
Hakimi H, Yamagishi J, Kawazu SI, Asada M. Advances in understanding red blood cell modifications by Babesia. PLoS Pathog 2022; 18:e1010770. [PMID: 36107982 PMCID: PMC9477259 DOI: 10.1371/journal.ppat.1010770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Babesia are tick-borne protozoan parasites that can infect livestock, pets, wildlife animals, and humans. In the mammalian host, they invade and multiply within red blood cells (RBCs). To support their development as obligate intracellular parasites, Babesia export numerous proteins to modify the RBC during invasion and development. Such exported proteins are likely important for parasite survival and pathogenicity and thus represent candidate drug or vaccine targets. The availability of complete genome sequences and the establishment of transfection systems for several Babesia species have aided the identification and functional characterization of exported proteins. Here, we review exported Babesia proteins; discuss their functions in the context of immune evasion, cytoadhesion, and nutrient uptake; and highlight possible future topics for research and application in this field.
Collapse
Affiliation(s)
- Hassan Hakimi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
- * E-mail: (HH); (MA)
| | - Junya Yamagishi
- Division of Collaboration and Education, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Shin-ichiro Kawazu
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Masahito Asada
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
- * E-mail: (HH); (MA)
| |
Collapse
|
16
|
Introini V, Govendir MA, Rayner JC, Cicuta P, Bernabeu M. Biophysical Tools and Concepts Enable Understanding of Asexual Blood Stage Malaria. Front Cell Infect Microbiol 2022; 12:908241. [PMID: 35711656 PMCID: PMC9192966 DOI: 10.3389/fcimb.2022.908241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 04/27/2022] [Indexed: 12/02/2022] Open
Abstract
Forces and mechanical properties of cells and tissues set constraints on biological functions, and are key determinants of human physiology. Changes in cell mechanics may arise from disease, or directly contribute to pathogenesis. Malaria gives many striking examples. Plasmodium parasites, the causative agents of malaria, are single-celled organisms that cannot survive outside their hosts; thus, thost-pathogen interactions are fundamental for parasite’s biological success and to the host response to infection. These interactions are often combinations of biochemical and mechanical factors, but most research focuses on the molecular side. However, Plasmodium infection of human red blood cells leads to changes in their mechanical properties, which has a crucial impact on disease pathogenesis because of the interaction of infected red blood cells with other human tissues through various adhesion mechanisms, which can be probed and modelled with biophysical techniques. Recently, natural polymorphisms affecting red blood cell biomechanics have also been shown to protect human populations, highlighting the potential of understanding biomechanical factors to inform future vaccines and drug development. Here we review biophysical techniques that have revealed new aspects of Plasmodium falciparum invasion of red blood cells and cytoadhesion of infected cells to the host vasculature. These mechanisms occur differently across Plasmodium species and are linked to malaria pathogenesis. We highlight promising techniques from the fields of bioengineering, immunomechanics, and soft matter physics that could be beneficial for studying malaria. Some approaches might also be applied to other phases of the malaria lifecycle and to apicomplexan infections with complex host-pathogen interactions.
Collapse
Affiliation(s)
- Viola Introini
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Viola Introini,
| | - Matt A. Govendir
- European Molecular Biology Laboratory (EMBL) Barcelona, Barcelona, Spain
| | - Julian C. Rayner
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Pietro Cicuta
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Maria Bernabeu
- European Molecular Biology Laboratory (EMBL) Barcelona, Barcelona, Spain
| |
Collapse
|
17
|
Paoletta MS, Wilkowsky SE. Thrombospondin Related Anonymous Protein Superfamily in Vector-Borne Apicomplexans: The Parasite’s Toolkit for Cell Invasion. Front Cell Infect Microbiol 2022; 12:831592. [PMID: 35463644 PMCID: PMC9019593 DOI: 10.3389/fcimb.2022.831592] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/07/2022] [Indexed: 11/23/2022] Open
Abstract
Apicomplexan parasites transmitted by vectors, including Babesia spp. and Plasmodium spp., cause severe disease in both humans and animals. These parasites have a complex life cycle during which they migrate, invade, and replicate in contrasting hosts such as the mammal and the invertebrate vector. The interaction of parasites with the host cell is mediated by adhesive proteins which play a key role in the different cellular processes regarding successful progression of the life cycle. Thrombospondin related anonymous protein (TRAP) is a superfamily of adhesins that are involved in motility, invasion and egress of the parasite. These proteins are stored and released from apical organelles and have either one or two types of adhesive domains, namely thrombospondin type 1 repeat and von Willebrand factor type A, that upon secretion are located in the extracellular portion of the molecule. Proteins from the TRAP superfamily have been intensively studied in Plasmodium species and to a lesser extent in Babesia spp., where they have proven to be functionally relevant throughout the entire parasite’s journey both in the arthropod vector and in the mammalian host. In recent years new findings provided answers to the role of TRAP proteins and in some cases the function of these adhesins during the parasite’s life cycle was redefined. In this review we will discuss the current knowledge of the diverse roles of the TRAP superfamily in vector-borne parasites from Class Aconoidasida. We will focus on the varied approaches that allowed the understanding of protein function and the relevance of TRAP- superfamily throughout the entire parasite’s cell cycle.
Collapse
|
18
|
Song P, Cai YC, Chen MX, Chen SH, Chen JX. Enhanced phosphatidylserine exposure and erythropoiesis in Babesia microti-infected mice. Front Microbiol 2022; 13:1083467. [PMID: 36687590 PMCID: PMC9846230 DOI: 10.3389/fmicb.2023.1083467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/08/2022] [Indexed: 01/05/2023] Open
Abstract
Introduction Babesia microti (B. microti) is the dominant species responsible for human babesiosis, which is associated with severe hemolytic anemia and splenomegaly because it infects mammalian erythrocytes. The actual prevalence of B. microti is thought to have been substantially underestimated. Methods In this study, Bagg's albino/c (BALB/c) mice were intraperitoneally injected with B. microti-infected erythrocytes, and parasitemia was subsequently measured by calculating the proportion of infected erythrocytes. The ultrastructure of infected erythrocytes was observed using scanning and transmission electron microscopes. Quantifying phosphatidylserine (PS) exposure, oxidative stress, intracellular Ca2+, and erythropoiesis of erythrocytes were done using flow cytometry. The physiological indicators were analyzed using a Mindray BC-5000 Vet automatic hematology analyzer. Results Of note, 40.7 ± 5.9% of erythrocytes changed their structure and shrunk in the B. microti-infected group. The percentage of annexin V-positive erythrocytes and the levels of reactive oxygen species (ROS) in the erythrocytes were higher in the B. microti-infected group than in the control group at 10 dpi. Significant splenomegaly and severe anemia were also observed following B. microti infection. The parasitemia level in the B. microti-infected splenectomized group was higher than that of the B. microti-infected sham group. The population of early erythroblasts increased, and the late erythroblasts decreased in both the bone marrow and spleen tissues of the B. microti-infected group at 10 dpi. Discussion PS exposure and elevated ROS activities were hallmarks of eryptosis in the B. microti-infected group. This study revealed for the first time that B. microti could also induce eryptosis. At the higher parasitemia phase, the occurrence of severe anemia and significant changes in the abundance of erythroblasts in B. microti-infected mice group were established. The spleen plays a critical protective role in controlling B. microti infection and preventing anemia. B. microti infection could cause a massive loss of late erythroblasts and induce erythropoiesis.
Collapse
Affiliation(s)
- Peng Song
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China.,NHC Key Laboratory of Parasite and Vector Biology, Ministry of Public Health, Shanghai, China.,WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China.,Hainan Tropical Diseases Research Center (Chinese Center for Tropical Diseases Research, Hainan), Haikou, Hainan, China
| | - Yu-Chun Cai
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China.,NHC Key Laboratory of Parasite and Vector Biology, Ministry of Public Health, Shanghai, China.,WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China
| | - Mu-Xin Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China.,NHC Key Laboratory of Parasite and Vector Biology, Ministry of Public Health, Shanghai, China.,WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China.,Hainan Tropical Diseases Research Center (Chinese Center for Tropical Diseases Research, Hainan), Haikou, Hainan, China
| | - Shao-Hong Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China.,NHC Key Laboratory of Parasite and Vector Biology, Ministry of Public Health, Shanghai, China.,WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China
| | - Jia-Xu Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China.,NHC Key Laboratory of Parasite and Vector Biology, Ministry of Public Health, Shanghai, China.,WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China.,Hainan Tropical Diseases Research Center (Chinese Center for Tropical Diseases Research, Hainan), Haikou, Hainan, China
| |
Collapse
|
19
|
Abstract
Plasmodium malaria parasites use a unique substrate-dependent locomotion, termed gliding motility, to migrate through tissues and invade cells. Previously, it was thought that the small labile invasive stages that invade erythrocytes, merozoites, use this motility solely to penetrate target erythrocytes. Here we reveal that merozoites use gliding motility for translocation across host cells prior to invasion. This forms an important preinvasion step that is powered by a conserved actomyosin motor and is regulated by a complex signaling pathway. This work broadens our understanding of the role of gliding motility and invasion in the blood and will have a significant impact on our understanding of blood stage host–pathogen interactions and parasite biology, with implications for interventions targeting erythrocyte invasion. Plasmodium malaria parasites are obligate intracellular protozoans that use a unique form of locomotion, termed gliding motility, to move through host tissues and invade cells. The process is substrate dependent and powered by an actomyosin motor that drives the posterior translocation of extracellular adhesins which, in turn, propel the parasite forward. Gliding motility is essential for tissue translocation in the sporozoite and ookinete stages; however, the short-lived erythrocyte-invading merozoite stage has never been observed to undergo gliding movement. Here we show Plasmodium merozoites possess the ability to undergo gliding motility in vitro and that this mechanism is likely an important precursor step for successful parasite invasion. We demonstrate that two human infective species, Plasmodium falciparum and Plasmodium knowlesi, have distinct merozoite motility profiles which may reflect distinct invasion strategies. Additionally, we develop and validate a higher throughput assay to evaluate the effects of genetic and pharmacological perturbations on both the molecular motor and the complex signaling cascade that regulates motility in merozoites. The discovery of merozoite motility provides a model to study the glideosome and adds a dimension for work aiming to develop treatments targeting the blood stage invasion pathways.
Collapse
|
20
|
Recent Advances in Molecular Genetic Tools for Babesia. Vet Sci 2021; 8:vetsci8100222. [PMID: 34679052 PMCID: PMC8541370 DOI: 10.3390/vetsci8100222] [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: 07/08/2021] [Revised: 09/15/2021] [Accepted: 10/02/2021] [Indexed: 11/21/2022] Open
Abstract
Development of in vitro culture and completion of genome sequencing of several Babesia parasites promoted the efforts to establish transfection systems for these parasites to dissect the gene functions. It has been more than a decade since the establishment of first transfection for Babesia bovis, the causative agent of bovine babesiosis. However, the number of genes that were targeted by genetic tools in Babesia parasites is limited. This is partially due to the low efficiencies of these methods. The recent adaptation of CRISPR/Cas9 for genome editing of Babesia bovis can accelerate the efforts for dissecting this parasite’s genome and extend the knowledge on biological aspects of erythrocytic and tick stages of Babesia. Additionally, glmS ribozyme as a conditional knockdown system is available that could be used for the characterization of essential genes. The development of high throughput genetic tools is needed to dissect the function of multigene families, targeting several genes in a specific pathway, and finally genome-wide identification of essential genes to find novel drug targets. In this review, we summarized the current tools that are available for Babesia and the genes that are being targeted by these tools. This may draw a perspective for the future development of genetic tools and pave the way for the identification of novel drugs or vaccine targets.
Collapse
|
21
|
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
|
22
|
Hakimi H, Asada M, Ishizaki T, Kawazu S. Isolation of viable Babesia bovis merozoites to study parasite invasion. Sci Rep 2021; 11:16959. [PMID: 34417510 PMCID: PMC8379152 DOI: 10.1038/s41598-021-96365-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 07/31/2021] [Indexed: 12/22/2022] Open
Abstract
Babesia parasite invades exclusively red blood cell (RBC) in mammalian host and induces alterations to host cell for survival. Despite the importance of Babesia in livestock industry and emerging cases in humans, their basic biology is hampered by lack of suitable biological tools. In this study, we aimed to develop a synchronization method for Babesia bovis which causes the most pathogenic form of bovine babesiosis. Initially, we used compound 2 (C2), a specific inhibitor of cyclic GMP-dependent protein kinase (PKG), and a derivative of C2, ML10. While both inhibitors were able to prevent B. bovis egress from RBC and increased percentage of binary forms, removal of inhibitors from culture did not result in a synchronized egress of parasites. Because using PKG inhibitors alone was not efficient to induce a synchronized culture, we isolated viable and invasive B. bovis merozoites and showed dynamics of merozoite invasion and development in RBCs. Using isolated merozoites we showed that BbVEAP, VESA1-export associated protein, is essential for parasite development in the RBC while has no significant role in invasion. Given the importance of invasion for the establishment of infection, this study paves the way for finding novel antigens to be used in control strategies against bovine babesiosis.
Collapse
Affiliation(s)
- Hassan Hakimi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, 080-8555, Japan. .,Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA.
| | - Masahito Asada
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, 080-8555, Japan
| | - Takahiro Ishizaki
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 852-8523, Japan
| | - Shinichiro Kawazu
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, 080-8555, Japan.
| |
Collapse
|
23
|
Wiser MF. Unique Endomembrane Systems and Virulence in Pathogenic Protozoa. Life (Basel) 2021; 11:life11080822. [PMID: 34440567 PMCID: PMC8401336 DOI: 10.3390/life11080822] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/10/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
Virulence in pathogenic protozoa is often tied to secretory processes such as the expression of adhesins on parasite surfaces or the secretion of proteases to assisted in tissue invasion and other proteins to avoid the immune system. This review is a broad overview of the endomembrane systems of pathogenic protozoa with a focus on Giardia, Trichomonas, Entamoeba, kinetoplastids, and apicomplexans. The focus is on unique features of these protozoa and how these features relate to virulence. In general, the basic elements of the endocytic and exocytic pathways are present in all protozoa. Some of these elements, especially the endosomal compartments, have been repurposed by the various species and quite often the repurposing is associated with virulence. The Apicomplexa exhibit the most unique endomembrane systems. This includes unique secretory organelles that play a central role in interactions between parasite and host and are involved in the invasion of host cells. Furthermore, as intracellular parasites, the apicomplexans extensively modify their host cells through the secretion of proteins and other material into the host cell. This includes a unique targeting motif for proteins destined for the host cell. Most notable among the apicomplexans is the malaria parasite, which extensively modifies and exports numerous proteins into the host erythrocyte. These modifications of the host erythrocyte include the formation of unique membranes and structures in the host erythrocyte cytoplasm and on the erythrocyte membrane. The transport of parasite proteins to the host erythrocyte involves several unique mechanisms and components, as well as the generation of compartments within the erythrocyte that participate in extraparasite trafficking.
Collapse
Affiliation(s)
- Mark F Wiser
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
24
|
Goodswen SJ, Kennedy PJ, Ellis JT. Applying Machine Learning to Predict the Exportome of Bovine and Canine Babesia Species That Cause Babesiosis. Pathogens 2021; 10:660. [PMID: 34071992 PMCID: PMC8226867 DOI: 10.3390/pathogens10060660] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 01/08/2023] Open
Abstract
Babesia infection of red blood cells can cause a severe disease called babesiosis in susceptible hosts. Bovine babesiosis causes global economic loss to the beef and dairy cattle industries, and canine babesiosis is considered a clinically significant disease. Potential therapeutic targets against bovine and canine babesiosis include members of the exportome, i.e., those proteins exported from the parasite into the host red blood cell. We developed three machine learning-derived methods (two novel and one adapted) to predict for every known Babesia bovis, Babesia bigemina, and Babesia canis protein the probability of being an exportome member. Two well-studied apicomplexan-related species, Plasmodium falciparum and Toxoplasma gondii, with extensive experimental evidence on their exportome or excreted/secreted proteins were used as important benchmarks for the three methods. Based on 10-fold cross validation and multiple train-validation-test splits of training data, we expect that over 90% of the predicted probabilities accurately provide a secretory or non-secretory indicator. Only laboratory testing can verify that predicted high exportome membership probabilities are creditable exportome indicators. However, the presented methods at least provide those proteins most worthy of laboratory validation and will ultimately save time and money.
Collapse
Affiliation(s)
- Stephen J. Goodswen
- School of Life Sciences, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia;
| | - Paul J. Kennedy
- School of Computer Science, Faculty of Engineering and Information Technology, Australian Artificial Intelligence Institute, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia;
| | - John T. Ellis
- School of Life Sciences, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia;
| |
Collapse
|
25
|
Nie Z, Ao Y, Wang S, Shu X, Li M, Zhan X, Yu L, An X, Sun Y, Guo J, Zhao Y, He L, Zhao J. Erythrocyte Adhesion of Merozoite Surface Antigen 2c1 Expressed During Extracellular Stages of Babesia orientalis. Front Immunol 2021; 12:623492. [PMID: 34079537 PMCID: PMC8165267 DOI: 10.3389/fimmu.2021.623492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/22/2021] [Indexed: 11/22/2022] Open
Abstract
Babesia orientalis, a major infectious agent of water buffalo hemolytic babesiosis, is transmitted by Rhipicephalus haemaphysaloides. However, no effective vaccine is available. Essential antigens that are involved in parasite invasion of host red blood cells (RBCs) are potential vaccine candidates. Therefore, the identification and the conduction of functional studies of essential antigens are highly desirable. Here, we evaluated the function of B. orientalis merozoite surface antigen 2c1 (BoMSA-2c1), which belongs to the variable merozoite surface antigen (VMSA) family in B. orientalis. We developed a polyclonal antiserum against the purified recombinant (r)BoMSA-2c1 protein. Immunofluorescence staining results showed that BoMSA-2c1 was expressed only on extracellular merozoites, whereas the antigen was undetectable in intracellular parasites. RBC binding assays suggested that BoMSA-2c1 specifically bound to buffalo erythrocytes. Cytoadherence assays using a eukaryotic expression system in vitro further verified the binding and inhibitory ability of BoMSA-2c1. We found that BoMSA-2c1 with a GPI domain was expressed on the surface of HEK293T cells that bound to water buffalo RBCs, and that the anti-rBoMSA2c1 antibody inhibited this binding. These results indicated that BoMSA-2c1 was involved in mediating initial binding to host erythrocytes of B. orientalis. Identification of the occurrence of binding early in the invasion process may facilitate understanding of the growth characteristics, and may help in formulating strategies for the prevention and control of this parasite.
Collapse
Affiliation(s)
- Zheng Nie
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
| | - Yangsiqi Ao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
| | - Sen Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
| | - Xiang Shu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
| | - Muxiao Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
| | - Xueyan Zhan
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
| | - Long Yu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
| | - Xiaomeng An
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
| | - Yali Sun
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
| | - Jiaying Guo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
| | - Yangnan Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China
| | - Lan He
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Junlong Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture of the People's Republic of China, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| |
Collapse
|
26
|
Abstract
Obligate intracellular malaria parasites reside within a vacuolar compartment generated during invasion which is the principal interface between pathogen and host. To subvert their host cell and support their metabolism, these parasites coordinate a range of transport activities at this membrane interface that are critically important to parasite survival and virulence, including nutrient import, waste efflux, effector protein export, and uptake of host cell cytosol. Here, we review our current understanding of the transport mechanisms acting at the malaria parasite vacuole during the blood and liver-stages of development with a particular focus on recent advances in our understanding of effector protein translocation into the host cell by the Plasmodium Translocon of EXported proteins (PTEX) and small molecule transport by the PTEX membrane-spanning pore EXP2. Comparison to Toxoplasma gondii and other related apicomplexans is provided to highlight how similar and divergent mechanisms are employed to fulfill analogous transport activities.
Collapse
Affiliation(s)
- Josh R. Beck
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, United States of America
| | - Chi-Min Ho
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| |
Collapse
|
27
|
Paoletta MS, Laughery JM, Arias LSL, Ortiz JMJ, Montenegro VN, Petrigh R, Ueti MW, Suarez CE, Farber MD, Wilkowsky SE. The key to egress? Babesia bovis perforin-like protein 1 (PLP1) with hemolytic capacity is required for blood stage replication and is involved in the exit of the parasite from the host cell. Int J Parasitol 2021; 51:643-658. [PMID: 33753093 DOI: 10.1016/j.ijpara.2020.12.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 11/27/2020] [Accepted: 12/03/2020] [Indexed: 12/18/2022]
Abstract
Bovine babesiosis is a tick-borne disease caused by apicomplexan parasites of the Babesia genus that represents a major constraint to livestock production worldwide. Currently available vaccines are based on live parasites which have archetypal limitations. Our goal is to identify candidate antigens so that new and effective vaccines against Babesia may be developed. The perforin-like protein (PLP) family has been identified as a key player in cell traversal and egress in related apicomplexans and it was also identified in Babesia, but its function in this parasite remains unknown. The aim of this work was to define the PLP family in Babesia and functionally characterize PLP1, a representative member of the family in Babesia bovis. Bioinformatic analyses demonstrate a variable number of plp genes (four to eight) in the genomes of six different Babesia spp. and conservation of the family members at the secondary and tertiary structure levels. We demonstrate here that Babesia PLPs contain the critical domains present in other apicomplexan PLPs to display the lytic capacity. We then focused on the functional characterization of PLP1 of B. bovis, both in vitro and in vivo. PLP1 is expressed and exposed to the host immune system during infection and has high hemolytic capacity under a wide range of conditions in vitro. A B. bovis plp1 knockout line displayed a decreased growth rate in vitro compared with the wild type strain and a peculiar phenotype consisting of multiple parasites within a single red blood cell, although at low frequency. This phenotype suggests that the lack of PLP1 has a negative impact on the mechanism of egression of the parasite and, therefore, on its capacity to proliferate. It is possible that PLP1 is associated with other proteins in the processes of invasion and egress, which were found to have redundant mechanisms in related apicomplexans. Future work will be focused on unravelling the network of proteins involved in these essential parasite functions.
Collapse
Affiliation(s)
- Martina Soledad Paoletta
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO) INTA - CONICET, De Los Reseros y Dr. Nicolás Repetto s/n, P.O. Box 25 (B1712WAA), Castelar, Buenos Aires, Argentina
| | - Jacob Michael Laughery
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA 99164, USA
| | - Ludmila Sol López Arias
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO) INTA - CONICET, De Los Reseros y Dr. Nicolás Repetto s/n, P.O. Box 25 (B1712WAA), Castelar, Buenos Aires, Argentina
| | - José Manuel Jaramillo Ortiz
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO) INTA - CONICET, De Los Reseros y Dr. Nicolás Repetto s/n, P.O. Box 25 (B1712WAA), Castelar, Buenos Aires, Argentina
| | - Valeria Noely Montenegro
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO) INTA - CONICET, De Los Reseros y Dr. Nicolás Repetto s/n, P.O. Box 25 (B1712WAA), Castelar, Buenos Aires, Argentina
| | - Romina Petrigh
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO) INTA - CONICET, De Los Reseros y Dr. Nicolás Repetto s/n, P.O. Box 25 (B1712WAA), Castelar, Buenos Aires, Argentina
| | - Massaro W Ueti
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA 99164, USA; Animal Disease Research Unit, USDA-ARS, Washington State University, 3003 ADBF, P.O. Box 646630, Pullman, WA 99164, USA
| | - Carlos Esteban Suarez
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA 99164, USA; Animal Disease Research Unit, USDA-ARS, Washington State University, 3003 ADBF, P.O. Box 646630, Pullman, WA 99164, USA
| | - Marisa Diana Farber
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO) INTA - CONICET, De Los Reseros y Dr. Nicolás Repetto s/n, P.O. Box 25 (B1712WAA), Castelar, Buenos Aires, Argentina
| | - Silvina Elizabeth Wilkowsky
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO) INTA - CONICET, De Los Reseros y Dr. Nicolás Repetto s/n, P.O. Box 25 (B1712WAA), Castelar, Buenos Aires, Argentina.
| |
Collapse
|
28
|
Hakimi H, Templeton TJ, Sakaguchi M, Yamagishi J, Miyazaki S, Yahata K, Uchihashi T, Kawazu SI, Kaneko O, Asada M. Novel Babesia bovis exported proteins that modify properties of infected red blood cells. PLoS Pathog 2020; 16:e1008917. [PMID: 33017449 PMCID: PMC7561165 DOI: 10.1371/journal.ppat.1008917] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 10/15/2020] [Accepted: 08/20/2020] [Indexed: 11/19/2022] Open
Abstract
Babesia bovis causes a pathogenic form of babesiosis in cattle. Following invasion of red blood cells (RBCs) the parasite extensively modifies host cell structural and mechanical properties via the export of numerous proteins. Despite their crucial role in virulence and pathogenesis, such proteins have not been comprehensively characterized in B. bovis. Here we describe the surface biotinylation of infected RBCs (iRBCs), followed by proteomic analysis. We describe a multigene family (mtm) that encodes predicted multi-transmembrane integral membrane proteins which are exported and expressed on the surface of iRBCs. One mtm gene was downregulated in blasticidin-S (BS) resistant parasites, suggesting an association with BS uptake. Induced knockdown of a novel exported protein encoded by BBOV_III004280, named VESA export-associated protein (BbVEAP), resulted in a decreased growth rate, reduced RBC surface ridge numbers, mis-localized VESA1, and abrogated cytoadhesion to endothelial cells, suggesting that BbVEAP is a novel virulence factor for B. bovis.
Collapse
Affiliation(s)
- Hassan Hakimi
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
- * E-mail: (HH); (MA)
| | - Thomas J. Templeton
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Miako Sakaguchi
- Central Laboratory, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Junya Yamagishi
- Division of Collaboration and Education, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Shinya Miyazaki
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Kazuhide Yahata
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | | | - Shin-ichiro Kawazu
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Osamu Kaneko
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Masahito Asada
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
- * E-mail: (HH); (MA)
| |
Collapse
|
29
|
Shu X, Guo J, Nie Z, Xia Y, He L, Zhao J. A novel 53 kDa protein (BoP53) in Babesia orientalis poses the immunoreactivity using the infection serum. Parasitol Int 2020; 78:102152. [PMID: 32512049 DOI: 10.1016/j.parint.2020.102152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 11/18/2022]
Abstract
Babesia orientalis (B. orientalis) is responsible for water buffalo babesiosis, which caused serious economic losses in the south of China. Although the invasion process has been roughly described, there are still some unknown molecules that have not yet been identified. Recently, an invasion-related protein BOV57 has been identified in the Babesia bovis. However, there is no report available about the gene in B. orientalis. B. orientalis P53 (BoP53) sequence was obtained by blast BOV57 sequence in B. orientalis genome database, and the full length of the BoP53 gene is 1599 bp. BoP53 gene was cloned into a pGEX-6P-1 expression vector and expressed as a GST-tag fusion protein. The tertiary structure of BoP53 was predicted with the I-TASSER software. The native BoP53 was identified from of B. orientalis lysate incubation with mouse antiserum against rBoP53. BoP53 as a novel identified protein promotes the study of B. orientalis, the reaction of rBoP53 with the serum of B. orientalis-infected water buffalo but not with healthy buffalo serum indicated its good antigenicity. It may be a candidate antigen for the diagnosis of B. orientalis infection.
Collapse
Affiliation(s)
- Xiang Shu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Animal Epidemical Disease and Infectious Zoonoses, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Jiaying Guo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Animal Epidemical Disease and Infectious Zoonoses, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Zheng Nie
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Animal Epidemical Disease and Infectious Zoonoses, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yingjun Xia
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Animal Epidemical Disease and Infectious Zoonoses, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Lan He
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Animal Epidemical Disease and Infectious Zoonoses, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Preventive Veterinary Medicine, Wuhan, Hubei 430070, China.
| | - Junlong Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Animal Epidemical Disease and Infectious Zoonoses, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Preventive Veterinary Medicine, Wuhan, Hubei 430070, China.
| |
Collapse
|
30
|
Milner DS, Ray LJ, Saxon EB, Lambert C, Till R, Fenton AK, Sockett RE. DivIVA Controls Progeny Morphology and Diverse ParA Proteins Regulate Cell Division or Gliding Motility in Bdellovibrio bacteriovorus. Front Microbiol 2020; 11:542. [PMID: 32373080 PMCID: PMC7186360 DOI: 10.3389/fmicb.2020.00542] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/12/2020] [Indexed: 01/12/2023] Open
Abstract
The predatory bacterium B. bacteriovorus grows and divides inside the periplasm of Gram-negative bacteria, forming a structure known as a bdelloplast. Cell division of predators inside the dead prey cell is not by binary fission but instead by synchronous division of a single elongated filamentous cell into odd or even numbers of progeny cells. Bdellovibrio replication and cell division processes are dependent on the finite level of nutrients available from inside the prey bacterium. The filamentous growth and division process of the predator maximizes the number of progeny produced by the finite nutrients in a way that binary fission could not. To learn more about such an unusual growth profile, we studied the role of DivIVA in the growing Bdellovibrio cell. This protein is well known for its link to polar cell growth and spore formation in Gram-positive bacteria, but little is known about its function in a predatory growth context. We show that DivIVA is expressed in the growing B. bacteriovorus cell and controls cell morphology during filamentous cell division, but not the number of progeny produced. Bacterial Two Hybrid (BTH) analysis shows DivIVA may interact with proteins that respond to metabolic indicators of amino-acid biosynthesis or changes in redox state. Such changes may be relevant signals to the predator, indicating the consumption of prey nutrients within the sealed bdelloplast environment. ParA, a chromosome segregation protein, also contributes to bacterial septation in many species. The B. bacteriovorus genome contains three ParA homologs; we identify a canonical ParAB pair required for predatory cell division and show a BTH interaction between a gene product encoded from the same operon as DivIVA with the canonical ParA. The remaining ParA proteins are both expressed in Bdellovibrio but are not required for predator cell division. Instead, one of these ParA proteins coordinates gliding motility, changing the frequency at which the cells reverse direction. Our work will prime further studies into how one bacterium can co-ordinate its cell division with the destruction of another bacterium that it dwells within.
Collapse
Affiliation(s)
- David S Milner
- Laboratory C15, Division of Infections, Immunity and Microbes, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Luke J Ray
- Laboratory C15, Division of Infections, Immunity and Microbes, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Emma B Saxon
- Laboratory C15, Division of Infections, Immunity and Microbes, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Carey Lambert
- Laboratory C15, Division of Infections, Immunity and Microbes, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Rob Till
- Laboratory C15, Division of Infections, Immunity and Microbes, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Andrew K Fenton
- Laboratory C15, Division of Infections, Immunity and Microbes, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Renee Elizabeth Sockett
- Laboratory C15, Division of Infections, Immunity and Microbes, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
31
|
The parasitophorous vacuole of the blood-stage malaria parasite. Nat Rev Microbiol 2020; 18:379-391. [PMID: 31980807 DOI: 10.1038/s41579-019-0321-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2019] [Indexed: 12/31/2022]
Abstract
The pathology of malaria is caused by infection of red blood cells with unicellular Plasmodium parasites. During blood-stage development, the parasite replicates within a membrane-bound parasitophorous vacuole. A central nexus for host-parasite interactions, this unique parasite shelter functions in nutrient acquisition, subcompartmentalization and the export of virulence factors, making its functional molecules attractive targets for the development of novel intervention strategies to combat the devastating impact of malaria. In this Review, we explore the origin, development, molecular composition and functions of the parasitophorous vacuole of Plasmodium blood stages. We also discuss the relevance of the malaria parasite's intravacuolar lifestyle for successful erythrocyte infection and provide perspectives for future research directions in parasitophorous vacuole biology.
Collapse
|
32
|
Bilgic HB, Hacilarlioglu S, Bakirci S, Kose O, Unlu AH, Aksulu A, Pekagirbas M, Ahmed J, Deschermeier C, Langley G, Karagenc T. Comparison of protectiveness of recombinant Babesia ovis apical membrane antigen 1 and B. ovis-infected cell line as vaccines against ovine babesiosis. Ticks Tick Borne Dis 2019; 11:101280. [PMID: 31506224 DOI: 10.1016/j.ttbdis.2019.101280] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 08/21/2019] [Accepted: 08/23/2019] [Indexed: 01/17/2023]
Abstract
Babesiosis is a disease complex caused by unicellular Babesia parasites and among them, malignant ovine babesiosis caused by B. ovis has a devastating economical impact on the small ruminant industry. The control of disease is mainly based on chemotherapy and preventing animals from tick infestation and to date no vaccine is available against ovine babesiosis. The requirement for vaccination against B. ovis infection in endemically unstable regions is necessary for implementation of effective disease control measures. The aim of the present study was to evaluate the effectiveness of different immunisation protocols against disease in sheep experimentally vaccinated with recombinant B. ovis apical membrane antigen-1 (rBoAMA-1) and/or live, a B. ovis-infected cell line. Sheep were divided into four experimental groups, plus a control group. Animals were immunised either with the B. ovis stabilate, or with rBoAMA-1, or with both rBoAMA-1 and the B. ovis stabilate. Western blots and ELISAs indicated that immunisation with rBoAMA-1 resulted in generation of a specific response against the recombinant protein, but the degree of antibody response did not correlate with the level of induced protection against challenge. The strongest immune response was induced in animals co-immunised with the live B. ovis stabilate plus rBoAMA-1. Both the hematological and parasitological findings indicated that this co-immunisation regimen has vaccine potential to limit losses incurred by ovine babesiosis in endemic countries.
Collapse
Affiliation(s)
- Huseyin Bilgin Bilgic
- Aydin Adnan Menderes University, Faculty of Veterinary Medicine, Department of Parasitology, 09016, Isikli/Aydin, Turkey.
| | - Selin Hacilarlioglu
- Aydin Adnan Menderes University, Faculty of Veterinary Medicine, Department of Parasitology, 09016, Isikli/Aydin, Turkey.
| | - Serkan Bakirci
- Aydin Adnan Menderes University, Faculty of Veterinary Medicine, Department of Parasitology, 09016, Isikli/Aydin, Turkey.
| | - Onur Kose
- Aydin Adnan Menderes University, Faculty of Veterinary Medicine, Department of Parasitology, 09016, Isikli/Aydin, Turkey; Burdur Mehmet Akif Ersoy University, Faculty of Veterinary Medicine, Department of Parasitology, 15030, Istiklal Yerleskesi, Burdur, Turkey.
| | - Ahmet Hakan Unlu
- Aydin Adnan Menderes University, Faculty of Veterinary Medicine, Department of Parasitology, 09016, Isikli/Aydin, Turkey; Van Yuzuncu Yil University, Vocational High School of Gevas, Department of Veterinary Medicine, Programme of Laboratorian and Veterinary Health, 65700, Van, Turkey.
| | - Ayca Aksulu
- Aydin Adnan Menderes University, Faculty of Veterinary Medicine, Department of Parasitology, 09016, Isikli/Aydin, Turkey.
| | - Metin Pekagirbas
- Aydin Adnan Menderes University, Faculty of Veterinary Medicine, Department of Parasitology, 09016, Isikli/Aydin, Turkey.
| | - Jabbar Ahmed
- Institue for Parasitology and Tropical Veterinary Medicine, Faculty of Veterinary Medicine, Free University of Berlin, Germany.
| | - Christina Deschermeier
- Diagnostics Development Laboratory, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.
| | - Gordon Langley
- Laboratoire de Biologie Comparative des Apicomplexes, Institut Cochin, Inserm U1016, Cnrs UMR 8104, Faculte de Medecine - Universite Paris Descartes, 27, rue du Faubourg-Saint-Jacques, 75014 Paris, France.
| | - Tulin Karagenc
- Aydin Adnan Menderes University, Faculty of Veterinary Medicine, Department of Parasitology, 09016, Isikli/Aydin, Turkey.
| |
Collapse
|
33
|
Allred DR. Variable and Variant Protein Multigene Families in Babesia bovis Persistence. Pathogens 2019; 8:pathogens8020076. [PMID: 31212587 PMCID: PMC6630957 DOI: 10.3390/pathogens8020076] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/03/2019] [Accepted: 06/07/2019] [Indexed: 11/16/2022] Open
Abstract
Cattle infected with Babesia bovis face a bifurcated fate: Either die of the severe acute infection, or survive and carry for many years a highly persistent but generally asymptomatic infection. In this review, the author describes known and potential contributions of three variable or highly variant multigene-encoded families of proteins to persistence in the bovine host, and the mechanisms by which variability arises among these families. Ramifications arising from this variability are discussed.
Collapse
Affiliation(s)
- David R Allred
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL 32611, USA.
- Emerging Pathogens Institute, University of Florida, Gainesville, FL 32611, USA.
- Genetics Institute, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
34
|
Suarez CE, Alzan HF, Silva MG, Rathinasamy V, Poole WA, Cooke BM. Unravelling the cellular and molecular pathogenesis of bovine babesiosis: is the sky the limit? Int J Parasitol 2019; 49:183-197. [PMID: 30690089 PMCID: PMC6988112 DOI: 10.1016/j.ijpara.2018.11.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/21/2018] [Accepted: 11/21/2018] [Indexed: 11/21/2022]
Abstract
The global impact of bovine babesiosis caused by the tick-borne apicomplexan parasites Babesia bovis, Babesia bigemina and Babesia divergens is vastly underappreciated. These parasites invade and multiply asexually in bovine red blood cells (RBCs), undergo sexual reproduction in their tick vectors (Rhipicephalus spp. for B. bovis and B. bigemina, and Ixodes ricinus for B. divergens) and have a trans-ovarial mode of transmission. Babesia parasites can cause acute and persistent infections to adult naïve cattle that can occur without evident clinical signs, but infections caused by B. bovis are associated with more severe disease and increased mortality, and are considered to be the most virulent agent of bovine babesiosis. In addition, babesiosis caused by B. divergens has an important zoonotic potential. The disease caused by B. bovis and B. bigemina can be controlled, at least in part, using therapeutic agents or vaccines comprising live-attenuated parasites, but these methods are limited in terms of their safety, ease of deployability and long-term efficacy, and improved control measures are urgently needed. In addition, expansion of tick habitats due to climate change and other rapidly changing environmental factors complicate efficient control of these parasites. While the ability to cause persistent infections facilitates transmission and persistence of the parasite in endemic regions, it also highlights their capacity to evade the host immune responses. Currently, the mechanisms of immune responses used by infected bovines to survive acute and chronic infections remain poorly understood, warranting further research. Similarly, molecular details on the processes leading to sexual reproduction and the development of tick-stage parasites are lacking, and such tick-specific molecules can be targets for control using alternative transmission blocking vaccines. In this review, we identify and examine key phases in the life-cycle of Babesia parasites, including dependence on a tick vector for transmission, sexual reproduction of the parasite in the midgut of the tick, parasite-dependent invasion and egression of bovine RBCs, the role of the spleen in the clearance of infected RBCs (IRBCs), and age-related disease resistance in cattle, as opportunities for developing improved control measures. The availability of integrated novel research approaches including "omics" (such as genomics, transcriptomics, and proteomics), gene modification, cytoadhesion assays, RBC invasion assays and methods for in vitro induction of sexual-stage parasites will accelerate our understanding of parasite vulnerabilities. Further, producing new knowledge on these vulnerabilities, as well as taking full advantage of existing knowledge, by filling important research gaps should result in the development of next-generation vaccines to control acute disease and parasite transmission. Creative and effective use of current and future technical and computational resources are needed, in the face of the numerous challenges imposed by these highly evolved parasites, for improving the control of this disease. Overall, bovine babesiosis is recognised as a global disease that imposes a serious burden on livestock production and human livelihood, but it largely remains a poorly controlled disease in many areas of the world. Recently, important progress has been made in our understanding of the basic biology and host-parasite interactions of Babesia parasites, yet a good deal of basic and translational research is still needed to achieve effective control of this important disease and to improve animal and human health.
Collapse
Affiliation(s)
- Carlos E Suarez
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, United States; Animal Disease Research Unit, Agricultural Research Service, USDA, WSU, Pullman, WA, United States.
| | - Heba F Alzan
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, United States; Parasitology and Animal Diseases Department, National Research Center, Dokki, Giza, Egypt
| | - Marta G Silva
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, United States; Animal Disease Research Unit, Agricultural Research Service, USDA, WSU, Pullman, WA, United States
| | - Vignesh Rathinasamy
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia
| | - William A Poole
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia
| | - Brian M Cooke
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia.
| |
Collapse
|
35
|
Kinetics of the invasion and egress processes of Babesia divergens, observed by time-lapse video microscopy. Sci Rep 2018; 8:14116. [PMID: 30237573 PMCID: PMC6148197 DOI: 10.1038/s41598-018-32349-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 08/31/2018] [Indexed: 11/11/2022] Open
Abstract
Based on confocal fluorescence and bright field video microscopy, we present detailed observations on the processes of invasion and egress of erythrocytes by the apicomplexan parasite Babesia divergens. Time-lapse images reveal numerous unexpected findings associated with the dynamics of B. divergens and its ability to manipulate the erythrocyte during both processes in its asexual cycle under in vitro conditions. Despite the speed at which these processes occur and the small size of the parasite, we capture infective merozoites moving vigorously and causing striking deformations in the erythrocyte’s plasma membrane during an active invasion. We also observed intraerythrocytic dynamic stages as paired pyriforms, double paired pyriforms, tetrads, unattached pyriform sister cells and multiple parasite stages resulting in the release of large numbers of merozoites over a short period. Of considerable interest is that time-lapse images reveal a novel mechanism of egress used by B. divergens to exit the human erythrocyte. The release occurs when B. divergens parasites establish contacts with the plasma membrane of the erythrocyte from within, before exiting the cell. Visualization and analysis of the images enabled us to obtain useful information and broaden our knowledge of complex and crucial events involved with parasitisation of human erythrocytes by B. divergens.
Collapse
|
36
|
Jalovecka M, Hajdusek O, Sojka D, Kopacek P, Malandrin L. The Complexity of Piroplasms Life Cycles. Front Cell Infect Microbiol 2018; 8:248. [PMID: 30083518 PMCID: PMC6065256 DOI: 10.3389/fcimb.2018.00248] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 06/29/2018] [Indexed: 01/23/2023] Open
Abstract
Although apicomplexan parasites of the group Piroplasmida represent commonly identified global risks to both animals and humans, detailed knowledge of their life cycles is surprisingly limited. Such a discrepancy results from incomplete literature reports, nomenclature disunity and recently, from large numbers of newly described species. This review intends to collate and summarize current knowledge with respect to piroplasm phylogeny. Moreover, it provides a comprehensive view of developmental events of Babesia, Theileria, and Cytauxzoon representative species, focusing on uniform consensus of three consecutive phases: (i) schizogony and merogony, asexual multiplication in blood cells of the vertebrate host; (ii) gamogony, sexual reproduction inside the tick midgut, later followed by invasion of kinetes into the tick internal tissues; and (iii) sporogony, asexual proliferation in tick salivary glands resulting in the formation of sporozoites. However, many fundamental differences in this general consensus occur and this review identifies variables that should be analyzed prior to further development of specific anti-piroplasm strategies, including the attractive targeting of life cycle stages of Babesia or Theileria tick vectors.
Collapse
Affiliation(s)
- Marie Jalovecka
- BIOEPAR, INRA, Oniris, Université Bretagne Loire, Nantes, France.,Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czechia.,Faculty of Science, University of South Bohemia, České Budějovice, Czechia
| | - Ondrej Hajdusek
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czechia
| | - Daniel Sojka
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czechia
| | - Petr Kopacek
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czechia
| | | |
Collapse
|
37
|
Abstract
Apicomplexa are obligate intracellular parasites that actively invade, replicate within, and egress from host cells. The parasite actinomyosin-based molecular motor complex (often referred to as the glideosome) is considered an important mediator of parasite motility and virulence. Mature intracellular parasites often become motile just prior to egress from their host cells, and in some genera, this motility is important for successful egress as well as for subsequent invasion of new host cells. To determine whether actinomyosin-based motility is important in the red blood cell egress and invasion activities of the malaria parasite, we have used a conditional genetic approach to delete GAP45, a primary component of the glideosome, in asexual blood stages of Plasmodium falciparum Our results confirm the essential nature of GAP45 for invasion but show that P. falciparum does not require a functional motor complex to undergo egress from the red blood cell. Malarial egress therefore differs fundamentally from induced egress in the related apicomplexan Toxoplasma gondiiIMPORTANCE Clinical malaria results from cycles of replication of single-celled parasites of the genus Plasmodium in red blood cells. Intracellular parasite replication is followed by a highly regulated, protease-dependent process called egress, in which rupture of the bounding membranes allows explosive release of daughter merozoites which rapidly invade fresh red cells. A parasite actinomyosin-based molecular motor (the glideosome) has been proposed to provide the mechanical force to drive invasion. Studies of the related parasite Toxoplasma gondii have shown that induced egress requires parasite motility, mediated by a functional glideosome. However, whether the glideosome has a similar essential role in egress of malaria merozoites from red blood cells is unknown. Here, we show that although a functional glideosome is required for red blood cell invasion by Plasmodium falciparum merozoites, it is not required for egress. These findings place further emphasis on the key role of the protease cascade in malarial egress.
Collapse
|
38
|
The effects of nitidine chloride and camptothecin on the growth of Babesia and Theileria parasites. Ticks Tick Borne Dis 2018; 9:1192-1201. [PMID: 29730263 DOI: 10.1016/j.ttbdis.2018.04.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/19/2018] [Accepted: 04/26/2018] [Indexed: 01/31/2023]
Abstract
The treatment of bovine and equine piroplasmosis is limited to diminazene aceturate (DA) and imidocarb dipropionate. To address this challenge, we need to explore novel drug compounds and targets. Topoisomerases are potential drug targets because they play a vital role in solving topological errors of DNA strands during replication. This study documented the effectiveness of topoisomerase inhibitors, nitidine chloride (NC) and camptothecin (Cpt), on the growth of Babesia and Theileria parasites. The half maximal inhibitory concentrations (IC50s) against B. bovis, B. bigemina, B. caballi, and T. equi were 1.01 ± 0.2, 5.34 ± 1.0, 0.11 ± 0.03, and 2.05 ± 0.4 μM for NC and 11.67 ± 1.6, 4.00 ± 1.0, 2.07 ± 0.6, and 0.33 ± 0.02 μM for Cpt, respectively. The viability experiment revealed that 4, 10, and 4 μM treatments of NC or 48, 8, and 8 μM treatments of Cpt were sufficient to stop the in vitro regrowth of B. bovis, B. bigemina, and B. caballi, respectively. However, T. equi regrew in all of the concentrations used. Moreover, increasing the concentration of NC and Cpt to 16 μM and 1.2 μM (8 × IC50) did not eliminate T. equi. The micrographs of B. bigemina and B. caballi taken at 24 h and 72 h showed deformed merozoites and remnants of parasites within the red blood cell (RBC), respectively. The treatments of 25 mg/kg DA and 20 mg/kg NC administered intraperitoneally and 20 mg/kg NC given orally showed 93.7, 90.7, and 83.6% inhibition against Babesia microti (B. microti), respectively, compared to the untreated group on day 8. In summary, NC and Cpt were effective against Babesia and Theileria parasites in vitro. Moreover, 20 mg/kg NC administered intraperitoneally was as effective as 25 mg/kg DA against B. microti in mice and showed no toxic symptoms in mice. The results indicate that NC may, after further evaluations, prove to be an alternative drug against bovine and equine piroplasmoses.
Collapse
|
39
|
SCUDIERO L, MERCADO-ROJANO WDEJ, RUDOLPH A, WANG J, LAUGHERY J, SUAREZ C. Comparisons of the topographic characteristics and electrical charge distributions amongBabesia-infected erythrocytes and extraerythrocytic merozoites using AFM. J Microsc 2018; 271:84-97. [DOI: 10.1111/jmi.12697] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/09/2018] [Accepted: 02/24/2018] [Indexed: 11/29/2022]
Affiliation(s)
- L. SCUDIERO
- Department of Chemistry and Materials Science and Engineering Program; Washington State University; Pullman Washington U.S.A
| | - W. DE J. MERCADO-ROJANO
- Department of Chemistry and Materials Science and Engineering Program; Washington State University; Pullman Washington U.S.A
| | - A. RUDOLPH
- Department of Chemistry and Materials Science and Engineering Program; Washington State University; Pullman Washington U.S.A
| | - J. WANG
- Department of Chemistry and Materials Science and Engineering Program; Washington State University; Pullman Washington U.S.A
| | - J.M. LAUGHERY
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology; Washington State University; Pullman Washington U.S.A
| | - C.E. SUAREZ
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology; Washington State University; Pullman Washington U.S.A
- Animal Disease Research Unit, Agricultural Research Service; United States Department of Agriculture; Pullman Washington U.S.A
| |
Collapse
|
40
|
Asada M, Hakimi H, Kawazu SI. The application of the HyPer fluorescent sensor in the real-time detection of H 2O 2 in Babesia bovis merozoites in vitro. Vet Parasitol 2018; 255:78-82. [PMID: 29773141 DOI: 10.1016/j.vetpar.2018.03.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/14/2018] [Accepted: 03/18/2018] [Indexed: 01/24/2023]
Abstract
In recent years, genetically encoded fluorescent probes have allowed a dramatic advancement in time-lapse imaging, enabling this imaging modality to be used to investigate intracellular events in several apicomplexan parasite species. In this study, we constructed a plasmid vector to stably express a genetically encoded H2O2 sensor probe called HyPer in Babesia bovis. The HyPer-transfected parasite population was successfully developed and subjected to a time-lapse imaging analysis under in vitro culture conditions. HyPer was capable of sensing an increasing H2O2 concentration in the parasite cells which was induced by the administration of paraquat as a superoxide donor. HyPer fluorescence co-staining with MitoTracker Red indicated the mitochondria as the major source of reactive oxygen species (ROS) in parasite cells. The fluctuating ROS dynamics in the parasite gliding toward, attaching to, and invading the target red blood cell was visualized and monitored in real time with the HyPer expressing parasite population. This is the first report to describe the application of the HyPer probe in an imaging analysis involving Babesia parasites. Hyper-expressing parasites can be widely utilized in studies to investigate the mechanisms of emergence and the reduction of oxidative stress, as well as the signal transduction in the parasite cells during host invasion and intercellular development.
Collapse
Affiliation(s)
- Masahito Asada
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto 1-12-4, Nagasaki, 852-8523, Japan; National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, 080-8555, Japan.
| | - Hassan Hakimi
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto 1-12-4, Nagasaki, 852-8523, Japan; National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, 080-8555, Japan.
| | - Shin-Ichiro Kawazu
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, 080-8555, Japan.
| |
Collapse
|
41
|
Sherling ES, van Ooij C. Host cell remodeling by pathogens: the exomembrane system in Plasmodium-infected erythrocytes. FEMS Microbiol Rev 2017; 40:701-21. [PMID: 27587718 PMCID: PMC5007283 DOI: 10.1093/femsre/fuw016] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2016] [Indexed: 12/22/2022] Open
Abstract
Malaria is caused by infection of erythrocytes by parasites of the genus Plasmodium. To survive inside erythrocytes, these parasites induce sweeping changes within the host cell, one of the most dramatic of which is the formation of multiple membranous compartments, collectively referred to as the exomembrane system. As an uninfected mammalian erythrocyte is devoid of internal membranes, the parasite must be the force and the source behind the formation of these compartments. Even though the first evidence of the presence these of internal compartments was obtained over a century ago, their functions remain mostly unclear, and in some cases completely unknown, and the mechanisms underlying their formation are still mysterious. In this review, we provide an overview of the different parts of the exomembrane system, describing the parasitophorous vacuole, the tubovesicular network, Maurer's clefts, the caveola-vesicle complex, J dots and other mobile compartments, and the small vesicles that have been observed in Plasmodium-infected cells. Finally, we combine the data into a simplified view of the exomembrane system and its relation to the alterations of the host erythrocyte. Plasmodium parasites remodel the host erythrocyte in various ways, including the formation of several membranous compartments, together referred to as the exomembrane system, within the erythrocyte cytosol that together are key to the sweeping changes in the host cell.
Collapse
Affiliation(s)
- Emma S Sherling
- The Francis Crick Institute, Mill Hill Laboratory, Mill Hill, London NW7 1AA, UK Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Christiaan van Ooij
- The Francis Crick Institute, Mill Hill Laboratory, Mill Hill, London NW7 1AA, UK
| |
Collapse
|
42
|
Ishizaki T, Sivakumar T, Hayashida K, Takemae H, Tuvshintulga B, Munkhjargal T, Guswanto A, Igarashi I, Yokoyama N. Babesia bovis BOV57, a Theileria parva P67 homolog, is an invasion-related, neutralization-sensitive antigen. INFECTION GENETICS AND EVOLUTION 2017; 54:138-145. [DOI: 10.1016/j.meegid.2017.06.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 06/19/2017] [Accepted: 06/25/2017] [Indexed: 11/27/2022]
|
43
|
Suarez CE, Bishop RP, Alzan HF, Poole WA, Cooke BM. Advances in the application of genetic manipulation methods to apicomplexan parasites. Int J Parasitol 2017; 47:701-710. [PMID: 28893636 DOI: 10.1016/j.ijpara.2017.08.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 08/24/2017] [Accepted: 08/24/2017] [Indexed: 12/13/2022]
Abstract
Apicomplexan parasites such as Babesia, Theileria, Eimeria, Cryptosporidium and Toxoplasma greatly impact animal health globally, and improved, cost-effective measures to control them are urgently required. These parasites have complex multi-stage life cycles including obligate intracellular stages. Major gaps in our understanding of the biology of these relatively poorly characterised parasites and the diseases they cause severely limit options for designing novel control methods. Here we review potentially important shared aspects of the biology of these parasites, such as cell invasion, host cell modification, and asexual and sexual reproduction, and explore the potential of the application of relatively well-established or newly emerging genetic manipulation methods, such as classical transfection or gene editing, respectively, for closing important gaps in our knowledge of the function of specific genes and proteins, and the biology of these parasites. In addition, genetic manipulation methods impact the development of novel methods of control of the diseases caused by these economically important parasites. Transient and stable transfection methods, in conjunction with whole and deep genome sequencing, were initially instrumental in improving our understanding of the molecular biology of apicomplexan parasites and paved the way for the application of the more recently developed gene editing methods. The increasingly efficient and more recently developed gene editing methods, in particular those based on the CRISPR/Cas9 system and previous conceptually similar techniques, are already contributing to additional gene function discovery using reverse genetics and related approaches. However, gene editing methods are only possible due to the increasing availability of in vitro culture, transfection, and genome sequencing and analysis techniques. We envisage that rapid progress in the development of novel gene editing techniques applied to apicomplexan parasites of veterinary interest will ultimately lead to the development of novel and more efficient methods for disease control.
Collapse
Affiliation(s)
- C E Suarez
- Animal Disease Research Unit, USDA-ARS, Washington State University, 3003 ADBF, P.O. Box 646630, Pullman, WA 99164, USA; Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA 99164, USA.
| | - R P Bishop
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA 99164, USA; The Paul G. Allen School for Global Animal Health, Washington State University, Pullman, WA, USA
| | - H F Alzan
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA 99164, USA; Parasitology and Animal Diseases Department, National Research Center, Dokki, Giza, Egypt
| | - W A Poole
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Victoria 3800, Australia
| | - B M Cooke
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Victoria 3800, Australia.
| |
Collapse
|
44
|
Eichenberger RM, Ramakrishnan C, Russo G, Deplazes P, Hehl AB. Genome-wide analysis of gene expression and protein secretion of Babesia canis during virulent infection identifies potential pathogenicity factors. Sci Rep 2017; 7:3357. [PMID: 28611446 PMCID: PMC5469757 DOI: 10.1038/s41598-017-03445-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 04/27/2017] [Indexed: 12/14/2022] Open
Abstract
Infections of dogs with virulent strains of Babesia canis are characterized by rapid onset and high mortality, comparable to complicated human malaria. As in other apicomplexan parasites, most Babesia virulence factors responsible for survival and pathogenicity are secreted to the host cell surface and beyond where they remodel and biochemically modify the infected cell interacting with host proteins in a very specific manner. Here, we investigated factors secreted by B. canis during acute infections in dogs and report on in silico predictions and experimental analysis of the parasite’s exportome. As a backdrop, we generated a fully annotated B. canis genome sequence of a virulent Hungarian field isolate (strain BcH-CHIPZ) underpinned by extensive genome-wide RNA-seq analysis. We find evidence for conserved factors in apicomplexan hemoparasites involved in immune-evasion (e.g. VESA-protein family), proteins secreted across the iRBC membrane into the host bloodstream (e.g. SA- and Bc28 protein families), potential moonlighting proteins (e.g. profilin and histones), and uncharacterized antigens present during acute crisis in dogs. The combined data provides a first predicted and partially validated set of potential virulence factors exported during fatal infections, which can be exploited for urgently needed innovative intervention strategies aimed at facilitating diagnosis and management of canine babesiosis.
Collapse
Affiliation(s)
| | | | | | - Peter Deplazes
- Institute of Parasitology, University of Zurich, Zurich, Switzerland
| | - Adrian B Hehl
- Institute of Parasitology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
45
|
Tardieux I, Baum J. Reassessing the mechanics of parasite motility and host-cell invasion. J Cell Biol 2017; 214:507-15. [PMID: 27573462 PMCID: PMC5004448 DOI: 10.1083/jcb.201605100] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 08/09/2016] [Indexed: 12/20/2022] Open
Abstract
The capacity to migrate is fundamental to multicellular and single-celled life. Apicomplexan parasites, an ancient protozoan clade that includes malaria parasites (Plasmodium) and Toxoplasma, achieve remarkable speeds of directional cell movement. This rapidity is achieved via a divergent actomyosin motor system, housed within a narrow compartment that lies underneath the length of the parasite plasma membrane. How this motor functions at a mechanistic level during motility and host cell invasion is a matter of debate. Here, we integrate old and new insights toward refining the current model for the function of this motor with the aim of revitalizing interest in the mechanics of how these deadly pathogens move.
Collapse
Affiliation(s)
- Isabelle Tardieux
- Institute of Advanced BioSciences, Institut National de la Santé et de la Recherche Médicale U1209, Centre National de la Recherche Scientifique UMR 5309, Université Grenoble Alpes, 38000, Grenoble, France
| | - Jake Baum
- Department of Life Sciences, Imperial College London, London SW7 2AZ, England, UK
| |
Collapse
|
46
|
Oldiges DP, Laughery JM, Tagliari NJ, Leite Filho RV, Davis WC, da Silva Vaz I, Termignoni C, Knowles DP, Suarez CE. Transfected Babesia bovis Expressing a Tick GST as a Live Vector Vaccine. PLoS Negl Trop Dis 2016; 10:e0005152. [PMID: 27911903 PMCID: PMC5135042 DOI: 10.1371/journal.pntd.0005152] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 11/01/2016] [Indexed: 11/18/2022] Open
Abstract
The Rhipicephalus microplus tick is a notorious blood-feeding ectoparasite of livestock, especially cattle, responsible for massive losses in animal production. It is the main vector for transmission of pathogenic bacteria and parasites, including Babesia bovis, an intraerythrocytic apicomplexan protozoan parasite responsible for bovine Babesiosis. This study describes the development and testing of a live B. bovis vaccine expressing the protective tick antigen glutathione-S-transferase from Haemaphysalis longicornis (HlGST). The B. bovis S74-T3B parasites were electroporated with a plasmid containing the bidirectional Ef-1α (elongation factor 1 alpha) promoter of B. bovis controlling expression of two independent genes, the selectable marker GFP-BSD (green fluorescent protein–blasticidin deaminase), and HlGST fused to the MSA-1 (merozoite surface antigen 1) signal peptide from B. bovis. Electroporation followed by blasticidin selection resulted in the emergence of a mixed B. bovis transfected line (termed HlGST) in in vitro cultures, containing parasites with distinct patterns of insertion of both exogenous genes, either in or outside the Ef-1α locus. A B. bovis clonal line termed HlGST-Cln expressing intracellular GFP and HlGST in the surface of merozoites was then derived from the mixed parasite line HlGST using a fluorescent activated cell sorter. Two independent calf immunization trials were performed via intravenous inoculation of the HlGST-Cln and a previously described control consisting of an irrelevant transfected clonal line of B. bovis designated GFP-Cln. The control GFP-Cln line contains a copy of the GFP-BSD gene inserted into the Ef-1α locus of B. bovis in an identical fashion as the HIGST-Cln parasites. All animals inoculated with the HlGST-Cln and GFP-Cln transfected parasites developed mild babesiosis. Tick egg fertility and fully engorged female tick weight was reduced significantly in R. microplus feeding on HlGST-Cln-immunized calves. Collectively, these data show the efficacy of a transfected HlGST-Cln B. bovis parasite to induce detectable anti-glutathione-S-transferase antibodies and a reduction in tick size and fecundity of R. microplus feeding in experimentally inoculated animals. The cattle tick Rhipicephalus microplus is a hematophagous ectoparasite, responsible for the transmission of lethal parasites such as Babesia sp, limiting cattle production in tropical and subtropical regions of the world. There is an urgent emerging need for improved methods of control for these currently neglected tick and tick borne diseases. It is hypothesized that a dual attenuated-live vector vaccine containing a stably transfected tick antigen elicits protective immune responses against the parasite and the tick vector in vaccinated cattle. Live Babesia vaccines based on attenuated parasites are the only effective method available for preventing acute babesiosis. On the other hand, glutathione-S-transferase from Haemaphysalis longicornis (HlGST) is a known effective antigen against Rhipicephalus microplus, the most common vector for B. bovis. This study describes the development and testing of a transfected, B. bovis vaccine expressing HlGST against the tick R. microplus. A B. bovis clonal line designated HlGST-Cln expressing HlGST and GFP/BSD, and separately a control transfected B. bovis clonal line expressing only GFP/BSD was used to vaccinate calves in two independent experiments. All immunized calves developed mild babesiosis, and only calves immunized with the HlGST-Cln parasite line generated anti-HlGST antibodies. Tick egg fertility and fully engorged female tick weight were reduced significantly in R. microplus feeding on HlGST-Cln-vaccinated calves. Taken together, these data demonstrates the ability of transfected B. bovis to elicit antibodies against a heterologous tick antigen in cattle and to induce partial protection in the vaccinated animals against the cattle tick for the first time, representing a step toward the goal to produce a live vector anti-tick vaccine.
Collapse
Affiliation(s)
- Daiane P. Oldiges
- Centro de Biotecnologia Universidade Federal do Rio Grande do Sul, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Jacob M. Laughery
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, United States of America
| | - Nelson Junior Tagliari
- Faculdade de Veterinária Universidade Federal do Rio Grande do Sul; Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Ronaldo Viana Leite Filho
- Faculdade de Veterinária Universidade Federal do Rio Grande do Sul; Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - William C. Davis
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, United States of America
| | - Itabajara da Silva Vaz
- Centro de Biotecnologia Universidade Federal do Rio Grande do Sul, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Faculdade de Veterinária Universidade Federal do Rio Grande do Sul; Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Carlos Termignoni
- Centro de Biotecnologia Universidade Federal do Rio Grande do Sul, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Departamento de Bioquímica Universidade Federal do Rio Grande do Sul; Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Donald P. Knowles
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, United States of America
- Animal Disease Research Unit, Agricultural Research Service, United States Department of Agriculture, Pullman, Washington, United States of America
| | - Carlos E. Suarez
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, United States of America
- Animal Disease Research Unit, Agricultural Research Service, United States Department of Agriculture, Pullman, Washington, United States of America
- * E-mail: ,
| |
Collapse
|
47
|
Extensive Shared Chemosensitivity between Malaria and Babesiosis Blood-Stage Parasites. Antimicrob Agents Chemother 2016; 60:5059-63. [PMID: 27246780 DOI: 10.1128/aac.00928-16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 05/23/2016] [Indexed: 11/20/2022] Open
Abstract
The apicomplexan parasites that cause malaria and babesiosis invade and proliferate within erythrocytes. To assess the potential for common antiparasitic treatments, we measured the sensitivities of multiple species of Plasmodium and Babesia parasites to the chemically diverse collection of antimalarial compounds in the Malaria Box library. We observed that these parasites share sensitivities to a large fraction of the same inhibitors and we identified compounds with strong babesiacidal activity.
Collapse
|
48
|
Ebine K, Hirai M, Sakaguchi M, Yahata K, Kaneko O, Saito-Nakano Y. Plasmodium Rab5b is secreted to the cytoplasmic face of the tubovesicular network in infected red blood cells together with N-acylated adenylate kinase 2. Malar J 2016; 15:323. [PMID: 27316546 PMCID: PMC4912828 DOI: 10.1186/s12936-016-1377-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 06/08/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Rab5 GTPase regulates membrane trafficking between the plasma membrane and endosomes and harbours a conserved C-terminal isoprenyl modification that is necessary for membrane recruitment. Plasmodium falciparum encodes three Rab5 isotypes, and one of these, Rab5b (PfRab5b), lacks the C-terminal modification but possesses the N-terminal myristoylation motif. PfRab5b was reported to localize to the parasite periphery. However, the trafficking pathway regulated by PfRab5b is unknown. METHODS A complementation analysis of Rab5 isotypes was performed in Plasmodium berghei. A constitutively active PfRab5b mutant was expressed under the regulation of a ligand-dependent destabilization domain (DD)-tag system in P. falciparum. The localization of PfRab5b was evaluated after removing the ligand followed by selective permeabilization of the membrane with different detergents. Furthermore, P. falciparum N-terminally myristoylated adenylate kinase 2 (PfAK2) was co-expressed with PfRab5b, and trafficking of PfAK2 to the parasitophorous vacuole membrane was examined by confocal microscopy. RESULTS PfRab5b complemented the function of PbRab5b, however, the conventional C-terminally isoprenylated Rab5, PbRab5a or PbRab5c, did not. The constitutively active PfRab5b mutant localized to the cytosol of the parasite and the tubovesicular network (TVN), a region that extends from the parasitophorous vacuole membrane (PVM) in infected red blood cells (iRBCs). By removing the DD-ligand, parasite cytosolic PfRab5b signal disappeared and a punctate structure adjacent to the endoplasmic reticulum (ER) and parasite periphery accumulated. The peripheral PfRab5b was sensitive to extracellular proteolysis after treatment with streptolysin O, which selectively permeabilizes the red blood cell plasma membrane, indicating that PfRab5b localized on the iRBC cytoplasmic face of the TVN. Transport of PfAK2 to the PVM was abrogated by overexpression of PfRab5b, and PfAK2 accumulated in the punctate structure together with PfRab5b. CONCLUSION N-myristoylated Plasmodium Rab5b plays a role that is distinct from that of conventional mammalian Rab5 isotypes. PfRab5b localizes to a compartment close to the ER, translocated to the lumen of the organelle, and co-localizes with PfAK2. PfRab5b and PfAK2 are then transported to the TVN, and PfRab5b localizes on the iRBC cytoplasmic face of TVN. These data demonstrate that PfRab5b is transported from the parasite cytosol to TVN together with N-myristoylated PfAK2 via an uncharacterized membrane-trafficking pathway.
Collapse
Affiliation(s)
- Kazuo Ebine
- Department of Parasitology, National Institute of Infectious Diseases, Shinjuku-Ku, Tokyo, Japan. .,Division of Cellular Dynamics, National Institute for Basic Biology, Okazaki, Aichi, Japan.
| | - Makoto Hirai
- Department of Molecular and Cellular Parasitology, Graduate School of Medicine, Juntendo University, Bunkyo-Ku, Tokyo, Japan.,Department of Parasitology, Graduate School of Medicine, Gunma University, Gunma, Japan
| | - Miako Sakaguchi
- Central Laboratory, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Kazuhide Yahata
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Osamu Kaneko
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Yumiko Saito-Nakano
- Department of Parasitology, National Institute of Infectious Diseases, Shinjuku-Ku, Tokyo, Japan.
| |
Collapse
|
49
|
Characterization of a papain-like cysteine protease essential for the survival of Babesia ovis merozoites. Ticks Tick Borne Dis 2016; 7:85-93. [DOI: 10.1016/j.ttbdis.2015.09.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 08/11/2015] [Accepted: 09/02/2015] [Indexed: 11/15/2022]
|
50
|
Pellé KG, Jiang RHY, Mantel PY, Xiao YP, Hjelmqvist D, Gallego-Lopez GM, O T Lau A, Kang BH, Allred DR, Marti M. Shared elements of host-targeting pathways among apicomplexan parasites of differing lifestyles. Cell Microbiol 2015; 17:1618-39. [PMID: 25996544 DOI: 10.1111/cmi.12460] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 04/27/2015] [Accepted: 05/14/2015] [Indexed: 11/30/2022]
Abstract
Apicomplexans are a diverse group of obligate parasites occupying different intracellular niches that require modification to meet the needs of the parasite. To efficiently manipulate their environment, apicomplexans translocate numerous parasite proteins into the host cell. Whereas some parasites remain contained within a parasitophorous vacuole membrane (PVM) throughout their developmental cycle, others do not, a difference that affects the machinery needed for protein export. A signal-mediated pathway for protein export into the host cell has been characterized in Plasmodium parasites, which maintain the PVM. Here, we functionally demonstrate an analogous host-targeting pathway involving organellar staging prior to secretion in the related bovine parasite, Babesia bovis, a parasite that destroys the PVM shortly after invasion. Taking into account recent identification of a similar signal-mediated pathway in the coccidian parasite Toxoplasma gondii, we suggest a model in which this conserved pathway has evolved in multiple steps from signal-mediated trafficking to specific secretory organelles for controlled secretion to a complex protein translocation process across the PVM.
Collapse
Affiliation(s)
- Karell G Pellé
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA
| | - Rays H Y Jiang
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA.,The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Pierre-Yves Mantel
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA
| | - Yu-Ping Xiao
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL, USA
| | - Daisy Hjelmqvist
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA
| | - Gina M Gallego-Lopez
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Audrey O T Lau
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Byung-Ho Kang
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, USA
| | - David R Allred
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL, USA.,Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Matthias Marti
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA
| |
Collapse
|