1
|
Hentzschel F, Binder AM, Dorner LP, Herzel L, Nuglisch F, Sema M, Röver K, He B, Aguirre-Botero MC, Cyrklaff M, Funaya C, Frischknecht F. Microtubule inner proteins of Plasmodium are essential for transmission of malaria parasites. Proc Natl Acad Sci U S A 2025; 122:e2421737122. [PMID: 39908102 PMCID: PMC11831158 DOI: 10.1073/pnas.2421737122] [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: 10/21/2024] [Accepted: 11/25/2024] [Indexed: 02/07/2025] Open
Abstract
Microtubule inner proteins (MIPs) are microtubule-associated proteins that bind to tubulin from the luminal side. MIPs can be found in axonemes to stabilize flagellar beat or within cytoplasmic microtubules. Plasmodium spp. are the causative agents of malaria that feature different parasite forms across a complex life cycle with both unique and divergent microtubule-based arrays. Here, we investigate four MIPs in a rodent malaria parasite for their role in transmission to and from the mosquito. We show by single and double gene deletions that SPM1 and TrxL1, MIPs associated with subpellicular microtubules, are dispensable for transmission from the vertebrate host to the mosquito and back. In contrast, FAP20 and FAP52, MIPs associated with the axonemes of gametes, are essential for transmission to mosquitoes but only if both genes are deleted. In the absence of both FAP20 and FAP52, the B-tubule of the axoneme partly detaches from the A-tubule, resulting in the deficiency of axonemal beating and hence gamete formation and egress. Our data suggest that a high level of redundancy ensures microtubule stability in the transmissive stages of Plasmodium, which is important for parasite transmission.
Collapse
Affiliation(s)
- Franziska Hentzschel
- Heidelberg University Medical Faculty, Center for Infectious Diseases, Parasitology, Heidelberg69120, Germany
- German Center for Infection Research, Deutsches Zentrum for Infektionsforschung (DZIF), Partner Site Heidelberg69120, Germany
| | - Annika M. Binder
- Heidelberg University Medical Faculty, Center for Infectious Diseases, Parasitology, Heidelberg69120, Germany
| | - Lilian P. Dorner
- Heidelberg University Medical Faculty, Center for Infectious Diseases, Parasitology, Heidelberg69120, Germany
| | - Lea Herzel
- Heidelberg University Medical Faculty, Center for Infectious Diseases, Parasitology, Heidelberg69120, Germany
| | - Fenja Nuglisch
- Heidelberg University Medical Faculty, Center for Infectious Diseases, Parasitology, Heidelberg69120, Germany
| | - Meslo Sema
- Heidelberg University Medical Faculty, Center for Infectious Diseases, Parasitology, Heidelberg69120, Germany
- Department of Medical Laboratory Sciences, Debre Tabor University, Debre Tabor6300, Ethiopia
| | - Katharina Röver
- Heidelberg University Medical Faculty, Center for Infectious Diseases, Parasitology, Heidelberg69120, Germany
| | - Buyuan He
- Heidelberg University Medical Faculty, Center for Infectious Diseases, Parasitology, Heidelberg69120, Germany
| | - Manuela C. Aguirre-Botero
- Heidelberg University Medical Faculty, Center for Infectious Diseases, Parasitology, Heidelberg69120, Germany
- Department of Parasites and Insect Vectors, Institut Pasteur, Université Paris Cité, Malaria Infection and Immunity, BioSPC, Paris75015, France
| | - Marek Cyrklaff
- Heidelberg University Medical Faculty, Center for Infectious Diseases, Parasitology, Heidelberg69120, Germany
| | - Charlotta Funaya
- Electron Microscopy Core Facility, Heidelberg University, Heidelberg69120, Germany
| | - Friedrich Frischknecht
- Heidelberg University Medical Faculty, Center for Infectious Diseases, Parasitology, Heidelberg69120, Germany
- German Center for Infection Research, Deutsches Zentrum for Infektionsforschung (DZIF), Partner Site Heidelberg69120, Germany
| |
Collapse
|
2
|
Trickey ML, Chowdury M, Bramwell G, Counihan NA, de Koning-Ward TF. Utilisation of an in vivo malaria model to provide functional proof for RhopH1/CLAG essentiality and conserved orthology with P. falciparum. J Biomed Sci 2025; 32:13. [PMID: 39894870 PMCID: PMC11789411 DOI: 10.1186/s12929-024-01105-7] [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: 08/21/2024] [Accepted: 11/19/2024] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND Malaria parasites establish new permeation pathways (NPPs) at the red blood cell membrane to facilitate the transport of essential nutrients from the blood plasma into the infected host cell. The NPPs are critical to parasite survival and, therefore, in the pursuit of novel therapeutics are an attractive drug target. The NPPs of the human parasite, P. falciparum, have been linked to the RhopH complex, with the monoallelic paralogues clag3.1 and clag3.2 encoding the protein RhopH1/CLAG3 that likely forms the NPP channel-forming component. Yet curiously, the combined knockout of both clag3 genes does not completely eliminate NPP function. The essentiality of the clag3 genes is, however, complicated by three additional clag paralogs (clag2, clag8 and clag9) in P. falciparum that could also be contributing to NPP formation. METHODS Here, the rodent malaria species, P. berghei, was utilised to investigate clag essentiality since it contains only two clag genes, clagX and clag9. Allelic replacement of the regions encompassing the functional components of P. berghei clagX with either P. berghei clag9 or P. falciparum clag3.1 examined the relationship between the two P. berghei clag genes as well as functional orthology across the two species. An inducible P. berghei clagX knockout was created to examine the essentiality of the clag3 ortholog to both survival and NPP functionality. RESULTS It was revealed P. berghei CLAGX and CLAG9, which belong to two distinct phylogenetic clades, have separate non-complementary functions, and that clagX is the functional orthologue of P. falciparum clag3. The inducible clagX knockout in conjunction with a guanidinium chloride induced-haemolysis assay to assess NPP function provided the first evidence of CLAG essentiality to Plasmodium survival and NPP function in an in vivo model of infection. CONCLUSIONS This work provides valuable insight regarding the essentiality of the RhopH1 clag genes to the NPPs functionality and validates the continued investigation of the RhopH complex as a therapeutic target to treat malaria infections.
Collapse
Affiliation(s)
- Mitchell L Trickey
- School of Medicine, Deakin University, Geelong, Australia
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, Australia
| | - Mrittika Chowdury
- School of Medicine, Deakin University, Geelong, Australia
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, Australia
| | - Georgina Bramwell
- School of Life and Environmental Sciences, Deakin University, Geelong, Australia
| | - Natalie A Counihan
- School of Medicine, Deakin University, Geelong, Australia
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, Australia
| | - Tania F de Koning-Ward
- School of Medicine, Deakin University, Geelong, Australia.
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, Australia.
| |
Collapse
|
3
|
Das S, Unhale T, Marinach C, Valeriano Alegria BDC, Roux C, Madry H, Mohand Oumoussa B, Amino R, Iwanaga S, Briquet S, Silvie O. Constitutive expression of Cas9 and rapamycin-inducible Cre recombinase facilitates conditional genome editing in Plasmodium berghei. Sci Rep 2025; 15:2949. [PMID: 39849074 PMCID: PMC11758014 DOI: 10.1038/s41598-025-87114-4] [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: 10/15/2024] [Accepted: 01/16/2025] [Indexed: 01/25/2025] Open
Abstract
Malaria is caused by protozoan parasites of the genus Plasmodium and remains a global health concern. The parasite has a highly adaptable life cycle comprising successive rounds of asexual replication in a vertebrate host and sexual maturation in the mosquito vector Anopheles. Genetic manipulation of the parasite has been instrumental for deciphering the function of Plasmodium genes. Conventional reverse genetic tools cannot be used to study essential genes of the asexual blood stages, thereby necessitating the development of conditional strategies. Among various such strategies, the rapamycin-inducible dimerisable Cre (DiCre) recombinase system emerged as a powerful approach for conditional editing of essential genes in human-infecting P. falciparum and in the rodent malaria model parasite P. berghei. We previously generated a DiCre-expressing P. berghei line and validated it by conditionally deleting several essential asexual stage genes, revealing their important role also in sporozoites. Another potent tool is the CRISPR/Cas9 technology, which has enabled targeted genome editing with higher accuracy and specificity and greatly advanced genome engineering in Plasmodium spp. Here, we developed new P. berghei parasite lines by integrating the DiCre cassette and a fluorescent marker in parasites constitutively expressing Cas9. Owing to the dual integration of CRISPR/Cas9 and DiCre, these new lines allow unparalleled levels of gene modification and conditional regulation simultaneously. To illustrate the versatility of this new tool, we conditionally knocked out the essential gene encoding the claudin-like apicomplexan micronemal protein (CLAMP) in P. berghei and confirmed the role of CLAMP during invasion of erythrocytes.
Collapse
Affiliation(s)
- Samhita Das
- Sorbonne Université, CNRS, Inserm, Centre d'Immunologie et des Maladies Infectieuses, CIMI, F-75013 Paris, France
| | - Tanaya Unhale
- Sorbonne Université, CNRS, Inserm, Centre d'Immunologie et des Maladies Infectieuses, CIMI, F-75013 Paris, France
| | - Carine Marinach
- Sorbonne Université, CNRS, Inserm, Centre d'Immunologie et des Maladies Infectieuses, CIMI, F-75013 Paris, France
| | - Belsy Del Carmen Valeriano Alegria
- Sorbonne Université, CNRS, Inserm, Centre d'Immunologie et des Maladies Infectieuses, CIMI, F-75013 Paris, France
- Institut Pasteur, Laboratory of Ecology and Emergence of Arthropod-borne Pathogens, Paris, France
| | - Camille Roux
- Sorbonne Université, CNRS, Inserm, Centre d'Immunologie et des Maladies Infectieuses, CIMI, F-75013 Paris, France
| | - Hélène Madry
- Sorbonne Université, Inserm, Production et Analyse des données en Sciences de la vie et Santé, PASS, Plateforme Post-génomique de la Pitié-Salpêtrière, P3S, F-75005 Paris, France
| | - Badreddine Mohand Oumoussa
- Sorbonne Université, Inserm, Production et Analyse des données en Sciences de la vie et Santé, PASS, Plateforme Post-génomique de la Pitié-Salpêtrière, P3S, F-75005 Paris, France
| | - Rogerio Amino
- Institut Pasteur, Université Paris Cité, Malaria Infection and Immunity Unit, F-75015 Paris, France
| | - Shiroh Iwanaga
- Research Center for Infectious Disease Control, Department of Molecular Protozoology, Suita, Osaka 565-0871, Japan
| | - Sylvie Briquet
- Sorbonne Université, CNRS, Inserm, Centre d'Immunologie et des Maladies Infectieuses, CIMI, F-75013 Paris, France.
| | - Olivier Silvie
- Sorbonne Université, CNRS, Inserm, Centre d'Immunologie et des Maladies Infectieuses, CIMI, F-75013 Paris, France.
| |
Collapse
|
4
|
Bailey AJ, Ukegbu CV, Giorgalli M, Besson TRB, Christophides GK, Vlachou D. Intracellular Plasmodium aquaporin 2 is important for sporozoite production in the mosquito vector and malaria transmission. Proc Natl Acad Sci U S A 2023; 120:e2304339120. [PMID: 37883438 PMCID: PMC10622946 DOI: 10.1073/pnas.2304339120] [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: 03/15/2023] [Accepted: 09/09/2023] [Indexed: 10/28/2023] Open
Abstract
Malaria remains a devastating disease and, with current measures failing to control its transmission, there is a need for novel interventions. A family of proteins that have long been pursued as potential intervention targets are aquaporins, which are channels facilitating the movement of water and other solutes across membranes. We identify an aquaporin in malaria parasites and demonstrate that it is important for completion of Plasmodium development in the mosquito vector. Disruption of AQP2 in the human parasite Plasmodium falciparum and the rodent parasite Plasmodium berghei blocks sporozoite production inside oocysts established on mosquito midguts, greatly limiting parasite infection of salivary glands and transmission to a new host. In vivo epitope tagging of AQP2 in P. berghei, combined with immunofluorescence assays, reveals that the protein is localized in vesicle-like organelles found in the cytoplasm of gametocytes, ookinetes, and sporozoites. The number of these organelles varies between individual parasites and lifecycle stages suggesting that they are likely part of a dynamic endomembrane system. Phylogenetic analysis confirms that AQP2 is unique to malaria and closely related parasites and most closely resembles intracellular aquaporins. Structure prediction analyses identify several unusual features, including a large accessory extracellular loop and an arginine-to-phenylalanine substitution in the selectivity filter principally determining pore function, a unique feature among known aquaporins. This in conjunction with the importance of AQP2 for malaria transmission suggests that AQP2 may be a fruitful target of antimalarial interventions.
Collapse
Affiliation(s)
- Alexander J. Bailey
- Department of Life Sciences, Imperial College London, LondonSW7 2AZ, United Kingdom
| | | | - Maria Giorgalli
- Department of Life Sciences, Imperial College London, LondonSW7 2AZ, United Kingdom
| | | | | | - Dina Vlachou
- Department of Life Sciences, Imperial College London, LondonSW7 2AZ, United Kingdom
| |
Collapse
|
5
|
Ukegbu CV, Gomes AR, Giorgalli M, Campos M, Bailey AJ, Besson TRB, Billker O, Vlachou D, Christophides GK. Identification of genes required for Plasmodium gametocyte-to-sporozoite development in the mosquito vector. Cell Host Microbe 2023; 31:1539-1551.e6. [PMID: 37708854 DOI: 10.1016/j.chom.2023.08.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/03/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023]
Abstract
Malaria remains one of the most devastating infectious diseases. Reverse genetic screens offer a powerful approach to identify genes and molecular processes governing malaria parasite biology. However, the complex regulation of gene expression and genotype-phenotype associations in the mosquito vector, along with sexual reproduction, have hindered the development of screens in this critical part of the parasite life cycle. To address this, we developed a genetic approach in the rodent parasite Plasmodium berghei that, in combination with barcode sequencing, circumvents the fertilization roadblock and enables screening for gametocyte-expressed genes required for parasite infection of the mosquito Anopheles coluzzii. Our results confirm previous findings, validating our approach for scaling up, and identify genes necessary for mosquito midgut infection, oocyst development, and salivary gland infection. These findings can aid efforts to study malaria transmission biology and to develop interventions for controlling disease transmission.
Collapse
Affiliation(s)
| | - Ana Rita Gomes
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK; Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Maria Giorgalli
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Melina Campos
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Alexander J Bailey
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | | | - Oliver Billker
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton CB10 1SA, UK
| | - Dina Vlachou
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK.
| | | |
Collapse
|
6
|
Anand A, Chandana M, Ghosh S, Das R, Singh N, Vaishalli PM, Gantasala NP, Padmanaban G, Nagaraj VA. Significance of Plasmodium berghei Amino Acid Transporter 1 in Food Vacuole Functionality and Its Association with Cerebral Pathogenesis. Microbiol Spectr 2023; 11:e0494322. [PMID: 36976018 PMCID: PMC10101031 DOI: 10.1128/spectrum.04943-22] [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: 12/01/2022] [Accepted: 03/07/2023] [Indexed: 03/29/2023] Open
Abstract
The food vacuole plays a central role in the blood stage of parasite development by digesting host hemoglobin acquired from red blood cells and detoxifying the host heme released during hemoglobin digestion into hemozoin. Blood-stage parasites undergo periodic schizont bursts, releasing food vacuoles containing hemozoin. Clinical studies in malaria-infected patients and in vivo animal studies have shown the association of hemozoin with disease pathogenesis and abnormal host immune responses in malaria. Here, we perform a detailed in vivo characterization of putative Plasmodium berghei amino acid transporter 1 localized in the food vacuole to understand its significance in the malaria parasite. We show that the targeted deletion of amino acid transporter 1 in Plasmodium berghei leads to a swollen food vacuole phenotype with the accumulation of host hemoglobin-derived peptides. Plasmodium berghei amino acid transporter 1-knockout parasites produce less hemozoin, and the hemozoin crystals display a thin morphology compared with wild-type parasites. The knockout parasites show reduced sensitivity to chloroquine and amodiaquine by showing recrudescence. More importantly, mice infected with the knockout parasites are protected from cerebral malaria and display reduced neuronal inflammation and cerebral complications. Genetic complementation of the knockout parasites restores the food vacuole morphology with hemozoin levels similar to that of wild-type parasites, causing cerebral malaria in the infected mice. The knockout parasites also show a significant delay in male gametocyte exflagellation. Our findings highlight the significance of amino acid transporter 1 in food vacuole functionality and its association with malaria pathogenesis and gametocyte development. IMPORTANCE Food vacuoles of the malaria parasite are involved in the degradation of red blood cell hemoglobin. The amino acids derived from hemoglobin degradation support parasite growth, and the heme released is detoxified into hemozoin. Antimalarials such as quinolines target hemozoin formation in the food vacuole. Food vacuole transporters transport hemoglobin-derived amino acids and peptides from the food vacuole to the parasite cytosol. Such transporters are also associated with drug resistance. Here, we show that the deletion of amino acid transporter 1 in Plasmodium berghei leads to swollen food vacuoles with the accumulation of hemoglobin-derived peptides. The transporter-deleted parasites generate less hemozoin with thin crystal morphology and show reduced sensitivity to quinolines. Mice infected with transporter-deleted parasites are protected from cerebral malaria. There is also a delay in male gametocyte exflagellation, affecting transmission. Our findings uncover the functional significance of amino acid transporter 1 in the life cycle of the malaria parasite.
Collapse
Affiliation(s)
- Aditya Anand
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Manjunatha Chandana
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar, Odisha, India
| | - Sourav Ghosh
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Rahul Das
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Nalini Singh
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India
| | - Pradeep Mini Vaishalli
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | | | | | | |
Collapse
|
7
|
Creation and preclinical evaluation of genetically attenuated malaria parasites arresting growth late in the liver. NPJ Vaccines 2022; 7:139. [PMCID: PMC9636417 DOI: 10.1038/s41541-022-00558-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022] Open
Abstract
AbstractWhole-sporozoite (WSp) malaria vaccines induce protective immune responses in animal malaria models and in humans. A recent clinical trial with a WSp vaccine comprising genetically attenuated parasites (GAP) which arrest growth early in the liver (PfSPZ-GA1), showed that GAPs can be safely administered to humans and immunogenicity is comparable to radiation-attenuated PfSPZ Vaccine. GAPs that arrest late in the liver stage (LA-GAP) have potential for increased potency as shown in rodent malaria models. Here we describe the generation of four putative P. falciparum LA-GAPs, generated by CRISPR/Cas9-mediated gene deletion. One out of four gene-deletion mutants produced sporozoites in sufficient numbers for further preclinical evaluation. This mutant, PfΔmei2, lacking the mei2-like RNA gene, showed late liver growth arrest in human liver-chimeric mice with human erythrocytes, absence of unwanted genetic alterations and sensitivity to antimalarial drugs. These features of PfΔmei2 make it a promising vaccine candidate, supporting further clinical evaluation. PfΔmei2 (GA2) has passed regulatory approval for safety and efficacy testing in humans based on the findings reported in this study.
Collapse
|
8
|
Jia X, Liu F, Bai J, Zhang Y, Cui L, Cao Y, Luo E. Phosphatase inhibitors BVT-948 and alexidine dihydrochloride inhibit sexual development of the malaria parasite Plasmodium berghei. Int J Parasitol Drugs Drug Resist 2022; 19:81-88. [PMID: 35792443 PMCID: PMC9260261 DOI: 10.1016/j.ijpddr.2022.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/18/2022] [Accepted: 06/22/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND With the emergence of resistance to front-line antimalarials, there is an urgent need to develop new medicines, including those targeting sexual development. This study aimed to assess the activity of a panel of phosphatase inhibitors against the sexual development of Plasmodium berghei and evaluate their potential as transmission-blocking agents. METHODS Twenty-five compounds were screened for transmission-blocking activity in vitro using the P. berghei ookinete culture assay. The inhibitory effects on male gametogenesis, gamete-ookinete, and zygote-ookinete formation were evaluated. The transmission-blocking activity of two compounds was evaluated using an in vivo mosquito feeding assay. Their cytotoxic effects were assessed on the human cell line HepG2. RESULTS Twelve compounds inhibited P. berghei ookinete formation with an IC50 < 10 μM. Two compounds, BVT-948 and alexidine dihydrochloride, significantly inhibited different developmental stages from gametogenesis through ookinete maturation. They also showed a substantial in vivo transmission-blocking activity by the mosquito feeding assay. CONCLUSIONS Some phosphatase inhibitors effectively inhibited Plasmodium sexual development and exhibited evident transmission-blocking activity, suggesting that phosphatases are valid targets for antimalarial development.
Collapse
Affiliation(s)
- Xitong Jia
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110122, China
| | - Fei Liu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110122, China
| | - Jie Bai
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110122, China
| | - Yongzhe Zhang
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110122, China; Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Suite 304, Tampa, FL, 33612-9415, USA
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110122, China.
| | - Enjie Luo
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110122, China.
| |
Collapse
|
9
|
Kehrer J, Formaglio P, Muthinja JM, Weber S, Baltissen D, Lance C, Ripp J, Grech J, Meissner M, Funaya C, Amino R, Frischknecht F. Plasmodium
sporozoite disintegration during skin passage limits malaria parasite transmission. EMBO Rep 2022; 23:e54719. [PMID: 35403820 PMCID: PMC9253755 DOI: 10.15252/embr.202254719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 11/25/2022] Open
Abstract
During transmission of malaria‐causing parasites from mosquitoes to mammals, Plasmodium sporozoites migrate rapidly in the skin to search for a blood vessel. The high migratory speed and narrow passages taken by the parasites suggest considerable strain on the sporozoites to maintain their shape. Here, we show that the membrane‐associated protein, concavin, is important for the maintenance of the Plasmodium sporozoite shape inside salivary glands of mosquitoes and during migration in the skin. Concavin‐GFP localizes at the cytoplasmic periphery and concavin(−) sporozoites progressively round up upon entry of salivary glands. Rounded concavin(−) sporozoites fail to pass through the narrow salivary ducts and are rarely ejected by mosquitoes, while normally shaped concavin(−) sporozoites are transmitted. Strikingly, motile concavin(−) sporozoites disintegrate while migrating through the skin leading to parasite arrest or death and decreased transmission efficiency. Collectively, we suggest that concavin contributes to cell shape maintenance by riveting the plasma membrane to the subtending inner membrane complex. Interfering with cell shape maintenance pathways might hence provide a new strategy to prevent a malaria infection.
Collapse
Affiliation(s)
- Jessica Kehrer
- Integrative Parasitology Center for Infectious Diseases Heidelberg University Medical School Heidelberg Germany
- Infectious Diseases Imaging Platform Center for Infectious Diseases Heidelberg University Medical School Heidelberg Germany
| | - Pauline Formaglio
- Malaria Infection and Immunity Unit Department of Parasites and Insect Vectors Institut Pasteur Paris France
| | - Julianne Mendi Muthinja
- Integrative Parasitology Center for Infectious Diseases Heidelberg University Medical School Heidelberg Germany
| | - Sebastian Weber
- Electron Microscopy Core Facility Heidelberg University Heidelberg Germany
| | - Danny Baltissen
- Integrative Parasitology Center for Infectious Diseases Heidelberg University Medical School Heidelberg Germany
| | - Christopher Lance
- Integrative Parasitology Center for Infectious Diseases Heidelberg University Medical School Heidelberg Germany
| | - Johanna Ripp
- Integrative Parasitology Center for Infectious Diseases Heidelberg University Medical School Heidelberg Germany
| | - Janessa Grech
- Experimental Parasitology Ludwig Maximilian University Munich Planegg‐Martinsried Germany
| | - Markus Meissner
- Experimental Parasitology Ludwig Maximilian University Munich Planegg‐Martinsried Germany
| | - Charlotta Funaya
- Electron Microscopy Core Facility Heidelberg University Heidelberg Germany
| | - Rogerio Amino
- Malaria Infection and Immunity Unit Department of Parasites and Insect Vectors Institut Pasteur Paris France
| | - Friedrich Frischknecht
- Integrative Parasitology Center for Infectious Diseases Heidelberg University Medical School Heidelberg Germany
- German Center for Infection Research (DZIF), Partner Site Heidelberg Heidelberg Germany
| |
Collapse
|
10
|
Hirai M, Maeta A, Mori T, Mita T. Pb103 Regulates Zygote/Ookinete Development in Plasmodium berghei via Double Zinc Finger Domains. Pathogens 2021; 10:pathogens10121536. [PMID: 34959491 PMCID: PMC8707419 DOI: 10.3390/pathogens10121536] [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: 11/04/2021] [Revised: 11/22/2021] [Accepted: 11/22/2021] [Indexed: 11/25/2022] Open
Abstract
Sexual reproduction of Plasmodium parasites takes place in anopheline mosquitoes, where male and female gametes fuse to form zygotes and then ookinetes. These processes are orchestrated by stage-specific protein expression, which is mediated in part by translational repression. Accumulating evidence shows that RNA binding proteins (RBPs) play crucial roles in these processes. Here, we report the characterization of P. berghei 103 (Pb103), which encodes a protein possessing double zinc finger domains (ZFs), an RBP. Reporter parasites expressing azami green fluorescent protein (AGFP) under the endogenous Pb103 gene promoter (Pb103-AGFP reporter) showed that the AGFP fluorescent signal was detected from gametes to ookinetes, while AGFP mRNA was translationally repressed in female gametocytes. The Pb103-disrupted parasites (Pb103(−)) grew and produced gametocytes with similar efficiencies to those of wild-type parasites. However, no oocysts were formed in mosquitoes fed Pb103(−). An in vitro fertilization assay showed abortion at the zygote stage in Pb103(−), suggesting that Pb103 plays a critical role in zygote/ookinete development. Cross-fertilization assays with Pb103(−) and male- or female-sterile parasites revealed that Pb103 was essential exclusively for female gametes. To identify the domains critical for zygote/ookinete development, transgenic parasites expressing partially deleted Pb103 were generated and assayed for ookinete maturation. As a result, deleting either of two ZFs but not the C-terminal region abolished zygote/ookinete development, highlighting the indispensable roles of ZFs in parasite sexual development, most likely via translational repression.
Collapse
|
11
|
Beyer K, Kracht S, Kehrer J, Singer M, Klug D, Frischknecht F. Limited Plasmodium sporozoite gliding motility in the absence of TRAP family adhesins. Malar J 2021; 20:430. [PMID: 34717635 PMCID: PMC8557484 DOI: 10.1186/s12936-021-03960-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/19/2021] [Indexed: 12/01/2022] Open
Abstract
Background Plasmodium sporozoites are the highly motile forms of malaria-causing parasites that are transmitted by the mosquito to the vertebrate host. Sporozoites need to enter and cross several cellular and tissue barriers for which they employ a set of surface proteins. Three of these proteins are members of the thrombospondin related anonymous protein (TRAP) family. Here, potential additive, synergistic or antagonistic roles of these adhesion proteins were investigated. Methods Four transgenic Plasmodium berghei parasite lines that lacked two or all three of the TRAP family adhesins TRAP, TLP and TREP were generated using positive–negative selection. The parasite lines were investigated for their capacity to attach to and move on glass, their ability to egress from oocysts and their capacity to enter mosquito salivary glands. One strain was in addition interrogated for its capacity to infect mice. Results The major phenotype of the TRAP single gene deletion dominates additional gene deletion phenotypes. All parasite lines including the one lacking all three proteins were able to conduct some form of active, if unproductive movement. Conclusions The individual TRAP-family adhesins appear to play functionally distinct roles during motility and infection. Other proteins must contribute to substrate adhesion and gliding motility. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12936-021-03960-3.
Collapse
Affiliation(s)
- Konrad Beyer
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 344, 69120, Heidelberg, Germany
| | - Simon Kracht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 344, 69120, Heidelberg, Germany
| | - Jessica Kehrer
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 344, 69120, Heidelberg, Germany.,German Center for Infection Research, Partner Site Heidelberg, 69120, Heidelberg, Germany
| | - Mirko Singer
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 344, 69120, Heidelberg, Germany.,Experimental Parasitology, Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, Lena-Christ-Straße 48, Planegg, 82152, Munich, Germany
| | - Dennis Klug
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 344, 69120, Heidelberg, Germany.,Université de Strasbourg, CNRS UPR9022, INSERM U963, Institut de Biologie Moléculaire et Cellulaire, 67000, Strasbourg, France
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 344, 69120, Heidelberg, Germany. .,German Center for Infection Research, Partner Site Heidelberg, 69120, Heidelberg, Germany.
| |
Collapse
|
12
|
Kolli SK, Salman AM, Ramesar J, Chevalley-Maurel S, Kroeze H, Geurten FGA, Miyazaki S, Mukhopadhyay E, Marin-Mogollon C, Franke-Fayard B, Hill AVS, Janse CJ. Screening of viral-vectored P. falciparum pre-erythrocytic candidate vaccine antigens using chimeric rodent parasites. PLoS One 2021; 16:e0254498. [PMID: 34252120 PMCID: PMC8274855 DOI: 10.1371/journal.pone.0254498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/28/2021] [Indexed: 11/19/2022] Open
Abstract
To screen for additional vaccine candidate antigens of Plasmodium pre-erythrocytic stages, fourteen P. falciparum proteins were selected based on expression in sporozoites or their role in establishment of hepatocyte infection. For preclinical evaluation of immunogenicity of these proteins in mice, chimeric P. berghei sporozoites were created that express the P. falciparum proteins in sporozoites as an additional copy gene under control of the uis4 gene promoter. All fourteen chimeric parasites produced sporozoites but sporozoites of eight lines failed to establish a liver infection, indicating a negative impact of these P. falciparum proteins on sporozoite infectivity. Immunogenicity of the other six proteins (SPELD, ETRAMP10.3, SIAP2, SPATR, HT, RPL3) was analyzed by immunization of inbred BALB/c and outbred CD-1 mice with viral-vectored (ChAd63 or ChAdOx1, MVA) vaccines, followed by challenge with chimeric sporozoites. Protective immunogenicity was determined by analyzing parasite liver load and prepatent period of blood stage infection after challenge. Of the six proteins only SPELD immunized mice showed partial protection. We discuss both the low protective immunogenicity of these proteins in the chimeric rodent malaria challenge model and the negative effect on P. berghei sporozoite infectivity of several P. falciparum proteins expressed in the chimeric sporozoites.
Collapse
Affiliation(s)
- Surendra Kumar Kolli
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Ahmed M. Salman
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Jai Ramesar
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Hans Kroeze
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Fiona G. A. Geurten
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Shinya Miyazaki
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Ekta Mukhopadhyay
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | | - Adrian V. S. Hill
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Chris J. Janse
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
13
|
Aroonsri A, Kongsee J, Gunawan JD, Aubry DA, Shaw PJ. A cell-based ribozyme reporter system employing a chromosomally-integrated 5' exonuclease gene. BMC Mol Cell Biol 2021; 22:20. [PMID: 33726662 PMCID: PMC7967978 DOI: 10.1186/s12860-021-00357-7] [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: 06/26/2020] [Accepted: 02/28/2021] [Indexed: 11/10/2022] Open
Abstract
Background Bioinformatic genome surveys indicate that self-cleaving ribonucleic acids (ribozymes) appear to be widespread among all domains of life, although the functions of only a small number have been validated by biochemical methods. Alternatively, cell-based reporter gene assays can be used to validate ribozyme function. However, reporter activity can be confounded by phenomena unrelated to ribozyme-mediated cleavage of RNA. Results We established a ribozyme reporter system in Escherichia coli in which a significant reduction of reporter activity is manifest when an active ribozyme sequence is fused to the reporter gene and the expression of a foreign Bacillus subtilis RNaseJ1 5′ exonuclease is induced from a chromosomally-integrated gene in the same cell. Conclusions The reporter system could be useful for validating ribozyme function in candidate sequences identified from bioinformatics. Supplementary Information The online version contains supplementary material available at 10.1186/s12860-021-00357-7.
Collapse
Affiliation(s)
- Aiyada Aroonsri
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, 12120, Thailand.
| | - Jindaporn Kongsee
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, 12120, Thailand
| | - Jeremy David Gunawan
- School of Life Science, Indonesia International Institute for Life Sciences (i3L), Jakarta, 13210, Indonesia
| | - Daniel Abidin Aubry
- School of Life Science, Indonesia International Institute for Life Sciences (i3L), Jakarta, 13210, Indonesia
| | - Philip James Shaw
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, 12120, Thailand
| |
Collapse
|
14
|
Ukegbu CV, Christophides GK, Vlachou D. Identification of Three Novel Plasmodium Factors Involved in Ookinete to Oocyst Developmental Transition. Front Cell Infect Microbiol 2021; 11:634273. [PMID: 33791240 PMCID: PMC8005625 DOI: 10.3389/fcimb.2021.634273] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/10/2021] [Indexed: 12/11/2022] Open
Abstract
Plasmodium falciparum malaria remains a major cause of global morbidity and mortality, mainly in sub-Saharan Africa. The numbers of new malaria cases and deaths have been stable in the last years despite intense efforts for disease elimination, highlighting the need for new approaches to stop disease transmission. Further understanding of the parasite transmission biology could provide a framework for the development of such approaches. We phenotypically and functionally characterized three novel genes, PIMMS01, PIMMS57, and PIMMS22, using targeted disruption of their orthologs in the rodent parasite Plasmodium berghei. PIMMS01 and PIMMS57 are specifically and highly expressed in ookinetes, while PIMMS22 transcription starts already in gametocytes and peaks in sporozoites. All three genes show strong phenotypes associated with the ookinete to oocyst transition, as their disruption leads to very low numbers of oocysts and complete abolishment of transmission. PIMMS22 has a secondary essential function in the oocyst. Our results enrich the molecular understanding of the parasite-vector interactions and identify PIMMS01, PIMMS57, and PIMMS22 as new targets of transmission blocking interventions.
Collapse
Affiliation(s)
- Chiamaka V Ukegbu
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - George K Christophides
- Department of Life Sciences, Imperial College London, London, United Kingdom.,The Cyprus Institute, Nicosia, Cyprus
| | - Dina Vlachou
- Department of Life Sciences, Imperial College London, London, United Kingdom.,The Cyprus Institute, Nicosia, Cyprus
| |
Collapse
|
15
|
Gao W, Sun X, Li D, Sun L, He Y, Wei H, Jin F, Cao Y. Toll-like receptor 4, Toll-like receptor 7 and Toll-like receptor 9 agonists enhance immune responses against blood-stage Plasmodium chabaudi infection in BALB/c mice. Int Immunopharmacol 2020; 89:107096. [PMID: 33091818 DOI: 10.1016/j.intimp.2020.107096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/09/2020] [Accepted: 10/09/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Toll-like receptor (TLR) signals play vital roles during the blood-stage of malaria infections. However, the roles of TLR agonists in the regulation of immune responses and the development of protective immunity to malaria remain poorly understood. METHOD BALB/c mice were pre-treated with TLR4, TLR7 and TLR9 agonists, followed by infection with Plasmodium chabaudi. After infection, splenic dendritic cells (DCs), Th1 cells and programmed death-1 (PD-1) expressed on Th1 cells, as well as regulatory T cells (Tregs) were analyzed by flow cytometry. The levels of IFN-γ, TNF-α, TGF-β and IL-10 in splenocytes and IgG1 and IgG2a in serum were measured by ELISA. RESULT Administration of TLR4, TLR7 and TLR9 agonists prior to infection improved disease outcomes. All TLR agonists promoted DC activation, and the proportions of Th1 cells increased. In TLR4, TLR7 and TLR9 agonist treated groups the levels of pro-inflammatory cytokines IFN-γ and TNF-α were elevated, and IgG1 and IgG2a serum levels were also significantly increased. TLR4, TLR7 and TLR9 agonists diminished the activation of Tregs and down-regulated the anti-inflammatory cytokines TGF-β and IL-10. Finally, PD-1 expressed on Th1 cells were decreased in TLR4, TLR7 and TLR9 agonist treated groups compared with control groups. CONCLUSION TLR4, TLR7 and TLR9 agonists activated DC-mediated innate immune responses and adaptive immune response, which against the blood-stage of Plasmodium and might be applied to malaria protection and treatment.
Collapse
Affiliation(s)
- Wenyan Gao
- Department of Immunology, Basic Medicine College of China Medical University, Shenyang 110122, Liaoning, China; Department of Obstetrics, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| | - Xiaodan Sun
- Department of Immunology, Basic Medicine College of China Medical University, Shenyang 110122, Liaoning, China
| | - Danni Li
- Department of Immunology, Basic Medicine College of China Medical University, Shenyang 110122, Liaoning, China
| | - Lin Sun
- Department of Immunology, Basic Medicine College of China Medical University, Shenyang 110122, Liaoning, China
| | - Yang He
- Department of Immunology, Basic Medicine College of China Medical University, Shenyang 110122, Liaoning, China
| | - Huanping Wei
- Department of Immunology, Basic Medicine College of China Medical University, Shenyang 110122, Liaoning, China.
| | - Feng Jin
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| | - Yaming Cao
- Department of Immunology, Basic Medicine College of China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|
16
|
Improvement of CRISPR/Cas9 system by transfecting Cas9-expressing Plasmodium berghei with linear donor template. Commun Biol 2020; 3:426. [PMID: 32759952 PMCID: PMC7406498 DOI: 10.1038/s42003-020-01138-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 06/25/2020] [Indexed: 11/08/2022] Open
Abstract
Malaria is caused by infection with Plasmodium parasites and is a major public health concern. The CRISPR/Cas9 system is a promising technology, but still has technical problems, such as low efficiency and unexpected recombination. Here, we solved these problems by transfecting Cas9-expressing parasites with linear donor templates. The use of a linear donor template prevented unexpected recombination; in addition, constitutive expression of Cas9 enabled immediate cleavage of the target locus after transfection, allowing efficient integration of the donor template. Furthermore, due to the absence of the cNHEJ pathway, there were no off-target mutations in the resultant parasites. In addition, this developed method could be applied for multiple genetic modifications on different chromosomes and for large-scale chromosomal deletion in the subtelomeric region. Because of its robustness, high efficiency, and versatile applicability, we hope this method will be standard in the post-genomic era of Plasmodium species.
Collapse
|
17
|
Klug D, Goellner S, Kehrer J, Sattler J, Strauss L, Singer M, Lu C, Springer TA, Frischknecht F. Evolutionarily distant I domains can functionally replace the essential ligand-binding domain of Plasmodium TRAP. eLife 2020; 9:57572. [PMID: 32648541 PMCID: PMC7351488 DOI: 10.7554/elife.57572] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 06/25/2020] [Indexed: 02/02/2023] Open
Abstract
Inserted (I) domains function as ligand-binding domains in adhesins that support cell adhesion and migration in many eukaryotic phyla. These adhesins include integrin αβ heterodimers in metazoans and single subunit transmembrane proteins in apicomplexans such as TRAP in Plasmodium and MIC2 in Toxoplasma. Here we show that the I domain of TRAP is essential for sporozoite gliding motility, mosquito salivary gland invasion and mouse infection. Its replacement with the I domain from Toxoplasma MIC2 fully restores tissue invasion and parasite transmission, while replacement with the aX I domain from human integrins still partially restores liver infection. Mutations around the ligand binding site allowed salivary gland invasion but led to inefficient transmission to the rodent host. These results suggest that apicomplexan parasites appropriated polyspecific I domains in part for their ability to engage with multiple ligands and to provide traction for emigration into diverse organs in distant phyla. Malaria is an infectious disease caused by single-celled parasites known as Plasmodium. Humans and other animals with backbones – such as birds, reptiles and rodents – can become hosts for these parasites if an infected female mosquito feeds on their blood. Likewise, healthy mosquitoes can in turn become infected with Plasmodium if they feed on the blood of an infected animal. To complete their life cycle, Plasmodium parasites within a mosquito must become spore-like cells called sporozoites. These sporozoites are highly mobile and can get into the mosquitoes’ salivary glands, meaning they can be passed on to a new host when the insect feeds. During a mosquito bite the sporozoites are spat into the skin of the potential host, where they then need to migrate rapidly to enter the bloodstream. Once in the blood, the sporozoites can then get into liver cells and begin a new infection. One protein called TRAP, which is found on the surface of the sporozoites, is important for their migration and the infection of the salivary glands or liver. Yet it was not known how this happens at the level of the individual proteins involved. Klug et al. have now tested how a part of the TRAP protein, called the I domain, contributes to the infection process. In the experiments, the I domain of TRAP was deleted which showed that the sporozoites need this domain to be able to move around and get into the host tissues. Without the I domain the sporozoites were stuck and could not successfully infect either the mosquitoes, the livers of mice, or human liver cells grown in the laboratory. Klug et al. then replaced the Plasmodium I domain of TRAP with the I domain from a distantly related parasite called Toxoplasma gondii, which causes a condition known as toxoplasmosis. The I domain from Toxoplasma allowed the Plasmodium parasites to infect the host tissues again. This observation was unexpected because Toxoplasma and Plasmodium parasites have evolved separately over the last 800 million years and Toxoplasma does not infect insects. These findings suggest that the I domain of TRAP evolved to bind several other proteins in different tissues and hosts. Future studies will investigate which other parasite proteins TRAP works with to guide sporozoites to the salivary glands or liver. Knowledge of how these proteins act together may lead to new approaches for treating or preventing malaria. For example, some treatments could stop sporozoites from entering liver cells.
Collapse
Affiliation(s)
- Dennis Klug
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany.,Université de Strasbourg, CNRS UPR9022, INSERM U963, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Sarah Goellner
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany.,Department of Molecular Virology, Heidelberg University Medical School, Heidelberg, Germany
| | - Jessica Kehrer
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Julia Sattler
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Léanne Strauss
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Mirko Singer
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany.,Experimental Parasitology, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, München, Germany
| | - Chafen Lu
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, United States
| | - Timothy A Springer
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, United States
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| |
Collapse
|
18
|
Gao W, Sun X, Li D, Sun L, He Y, Wei H, Jin F, Cao Y. Toll-like receptor 7 and Toll-like receptor 9 agonists effectively enhance immunological memory in Plasmodium chabaudi infected BALB/c mice. Int Immunopharmacol 2020; 81:106248. [PMID: 32007799 DOI: 10.1016/j.intimp.2020.106248] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 01/22/2020] [Accepted: 01/22/2020] [Indexed: 11/29/2022]
Affiliation(s)
- Wenyan Gao
- Department of Immunology, Basic Medicine College of China Medical University, Shenyang 110122, Liaoning, China; Department of Obstetrics, The First Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Xiaodan Sun
- Department of Immunology, Basic Medicine College of China Medical University, Shenyang 110122, Liaoning, China
| | - Danni Li
- Department of Immunology, Basic Medicine College of China Medical University, Shenyang 110122, Liaoning, China
| | - Lin Sun
- Department of Immunology, Basic Medicine College of China Medical University, Shenyang 110122, Liaoning, China
| | - Yang He
- Department of Immunology, Basic Medicine College of China Medical University, Shenyang 110122, Liaoning, China
| | - Huanping Wei
- Department of Immunology, Basic Medicine College of China Medical University, Shenyang 110122, Liaoning, China
| | - Feng Jin
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Yaming Cao
- Department of Immunology, Basic Medicine College of China Medical University, Shenyang 110122, Liaoning, China.
| |
Collapse
|
19
|
Songsungthong W, Yongkiettrakul S, Bohan LE, Nicholson ES, Prasopporn S, Chaiyen P, Leartsakulpanich U. Diaminoquinazoline MMV675968 from Pathogen Box inhibits Acinetobacter baumannii growth through targeting of dihydrofolate reductase. Sci Rep 2019; 9:15625. [PMID: 31666629 PMCID: PMC6821926 DOI: 10.1038/s41598-019-52176-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/14/2019] [Indexed: 02/07/2023] Open
Abstract
Antibiotic resistance in Acinetobacter baumannii is a major global health threat. New drugs with novel chemical structures are needed to overcome a myriad of resistance mechanisms in A. baumannii. In this study, we screened an open-source Pathogen Box library for anti-A. baumannii compounds. Compound MMV675968 (a diaminoquinazoline analog) was the only non-reference compound found to inhibit the growth of all four A. baumannii test strains with IC50 of 0.6–2.7 μM, IC90 of 0.7–3.9 μM, and MIC of 1.6–10 μM. We showed that MMV675968 targeted A. baumannii dihydrofolate reductase (AbDHFR) as determined by an E. coli surrogate whose growth was dependent on AbDHFR function and by an in vitro DHFR activity assay. Additionally, chemical scaffolds of DHFR inhibitors that are effective as antibiotics against A. baumannii were identified using an in vitro DHFR activity assay and A. baumannii growth inhibition. MMV675968 was the most potent among DHFR inhibitors tested in inhibiting A. baumannii growth. This study shows for the first time that MMV675968 inhibits A. baumannii growth via selective inhibition of AbDHFR and is therefore a promising scaffold for further antibiotic development against A. baumannii.
Collapse
Affiliation(s)
- Warangkhana Songsungthong
- Biomolecular Analysis and Application Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), Pathum Thani, 12120, Thailand.
| | - Suganya Yongkiettrakul
- Biomolecular Analysis and Application Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), Pathum Thani, 12120, Thailand
| | - Louise E Bohan
- Biomolecular Analysis and Application Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), Pathum Thani, 12120, Thailand.,UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, 4, Ireland
| | - Eric S Nicholson
- Biomolecular Analysis and Application Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), Pathum Thani, 12120, Thailand.,Biology Department, Earlham College, Indiana, 47374, USA
| | - Sunisa Prasopporn
- Biomolecular Analysis and Application Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), Pathum Thani, 12120, Thailand.,Department of Pharmacology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Pimchai Chaiyen
- School of Biomolecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong, 21210, Thailand
| | - Ubolsree Leartsakulpanich
- Biomolecular Analysis and Application Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), Pathum Thani, 12120, Thailand
| |
Collapse
|
20
|
Liu F, Liu Q, Yu C, Zhao Y, Wu Y, Min H, Qiu Y, Jin Y, Miao J, Cui L, Cao Y. An MFS-Domain Protein Pb115 Plays a Critical Role in Gamete Fertilization of the Malaria Parasite Plasmodium berghei. Front Microbiol 2019; 10:2193. [PMID: 31616399 PMCID: PMC6764285 DOI: 10.3389/fmicb.2019.02193] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 09/06/2019] [Indexed: 12/20/2022] Open
Abstract
Sexual reproduction is an essential process in the Plasmodium life cycle and a vulnerable step for blocking transmission from the human host to mosquitoes. In this study, we characterized the functions of a conserved cell membrane protein P115 in the rodent malaria parasite Plasmodium berghei ANKA. Pb115 was expressed in both asexual stages (schizonts) and sexual stages (gametocytes, gametes, and ookinetes), and was localized on the plasma membrane of gametes and ookinetes. In P. berghei, genetic deletion of Pb115 (Δpb115) did not affect asexual multiplication, nor did it affect gametocyte development or exflagellation of the male gametocytes. However, mosquitoes fed on Δpb115-infected mice showed 74% reduction in the prevalence of infection and 96.5% reduction in oocyst density compared to those fed on wild-type P. berghei-infected mice. The Δpb115 parasites showed significant defects in the interactions between the male and female gametes, and as a result, very few zygotes were formed in ookinete cultures. Cross fertilization with the male-defective Δpbs48/45 line and the female-defective Δpfs47 line further indicated that the fertilization defects of the Δpb115 lines were present in both male and female gametes. We evaluated the transmission-blocking potential of Pb115 by immunization of mice with a recombinant Pb115 fragment. In vivo mosquito feeding assay showed Pb115 immunization conferred modest, but significant transmission reducing activity with 44% reduction in infection prevalence and 39% reduction in oocyst density. Our results described functional characterization of a conserved membrane protein as a fertility factor in Plasmodium and demonstrated transmission-blocking potential of this antigen.
Collapse
Affiliation(s)
- Fei Liu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Qingyang Liu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Chunyun Yu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yan Zhao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yudi Wu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Hui Min
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Tampa, FL, United States
| | - Yue Qiu
- The First Hospital of China Medical University, Shenyang, China
| | - Ying Jin
- Liaoning Research Institute of Family Planning, Shenyang, China
| | - Jun Miao
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Tampa, FL, United States
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Tampa, FL, United States
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| |
Collapse
|
21
|
Soga A, Shirozu T, Ko-Ketsu M, Fukumoto S. Improvement of an in vitro drug selection method for generating transgenic Plasmodium berghei parasites. Malar J 2019; 18:215. [PMID: 31238932 PMCID: PMC6593524 DOI: 10.1186/s12936-019-2851-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 06/19/2019] [Indexed: 11/20/2022] Open
Abstract
Background Reverse genetics approaches have become powerful tools to dissect the biology of malaria parasites. In a previous study, development of an in vitro drug selection method for generating transgenic parasite of Plasmodium berghei was reported. Using this method, two novel and independent selection markers using the P. berghei heat shock protein 70 promoter was previously established. While the approach permits the easy and flexible genetic manipulation of P. berghei, shortcomings include a low variety in promoter options to drive marker gene expression and increased complexity of the selection procedure. In this study, addressing these issues was attempted. Methods To secure a variety of promoters, the use of a P. berghei elongation factor-1α promoter for marker gene expression was attempted. To simplify the procedure of in vitro selection, the establishment of a two cell-cycle culture method and its application for drug selection were attempted. Results The P. berghei elongation factor-1α (pbef-1α) promoter, which is commonly used to drive marker gene expression, was successfully applied as an alternative promoter model for marker gene expression, using the parasite’s codon-optimized marker sequence. To simplify the in vitro selection method, a two cell-cycle culture method in which the merozoite was released by filtration of the culture containing matured schizont-infected erythrocytes was also developed and successfully applied for drug selection. Conclusion The pbef-1α promoter was successfully applied in an in vitro selection system. The in vitro selection procedure also could be simplified for practical use using a two cell-cycle culture method. These improvements provide a more versatile platform for the genetic manipulation of P. berghei.
Collapse
Affiliation(s)
- Akira Soga
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan
| | - Takahiro Shirozu
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan
| | - Mami Ko-Ketsu
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan
| | - Shinya Fukumoto
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan.
| |
Collapse
|
22
|
Obrova K, Cyrklaff M, Frank R, Mair GR, Mueller AK. Transmission of the malaria parasite requires ferlin for gamete egress from the red blood cell. Cell Microbiol 2019; 21:e12999. [PMID: 30597708 DOI: 10.1111/cmi.12999] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 11/15/2018] [Accepted: 12/09/2018] [Indexed: 02/02/2023]
Abstract
Ferlins mediate calcium-dependent vesicular fusion. Although conserved throughout eukaryotic evolution, their function in unicellular organisms including apicomplexan parasites is largely unknown. Here, we define a crucial role for a ferlin-like protein (FLP) in host-to-vector transmission of the rodent malaria parasite Plasmodium berghei. Infection of the mosquito vectors requires the formation of free gametes and their fertilisation in the mosquito midgut. Mature gametes will only emerge upon secretion of factors that stimulate the disruption of the red blood cell membrane and the parasitophorous vacuole membrane. Genetic depletion of FLP in sexual stages leads to a complete life cycle arrest in the mosquito. Although mature gametes form normally, mutants lacking FLP remain trapped in the red blood cell. The egress defect is rescued by detergent-mediated membrane lysis. In agreement with ferlin vesicular localisation, HA-tagged FLP labels intracellular speckles, which relocalise to the cell periphery during gamete maturation. Our data define FLP as a novel critical factor for Plasmodium fertilisation and transmission and suggest an evolutionarily conserved example of ferlin-mediated exocytosis.
Collapse
Affiliation(s)
- Klara Obrova
- Center for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Marek Cyrklaff
- Center for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Roland Frank
- Center for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Gunnar R Mair
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
| | - Ann-Kristin Mueller
- Center for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Infectious Diseases (DZIF), Universitätsklinikum Heidelberg, Heidelberg, Germany
| |
Collapse
|
23
|
Klug D, Kehrer J, Frischknecht F, Singer M. A synthetic promoter for multi-stage expression to probe complementary functions of Plasmodium adhesins. J Cell Sci 2018; 131:jcs.210971. [PMID: 30237220 DOI: 10.1242/jcs.210971] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 09/10/2018] [Indexed: 11/20/2022] Open
Abstract
Gene expression of malaria parasites is mediated by the apicomplexan Apetala2 (ApiAP2) transcription factor family. Different ApiAP2s control gene expression at distinct stages in the complex life cycle of the parasite, ensuring timely expression of stage-specific genes. ApiAP2s recognize short cis-regulatory elements that are enriched in the upstream/promoter region of their target genes. This should, in principle, allow the generation of 'synthetic' promoters that drive gene expression at desired stages of the Plasmodium life cycle. Here we test this concept by combining cis-regulatory elements of two genes expressed successively within the mosquito part of the life cycle. Our tailored 'synthetic' promoters, named Spooki 1.0 and Spooki 2.0, activate gene expression in early and late mosquito stages, as shown by the expression of a fluorescent reporter. We used these promoters to address the specific functionality of two related adhesins that are exclusively expressed either during the early or late mosquito stage. By modifying the expression profile of both adhesins in absence of their counterpart we were able to test for complementary functions in gliding and invasion. We discuss the possible advantages and drawbacks of our approach.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Dennis Klug
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Jessica Kehrer
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Mirko Singer
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| |
Collapse
|
24
|
The Glycosylphosphatidylinositol Transamidase Complex Subunit PbGPI16 of Plasmodium berghei Is Important for Inducing Experimental Cerebral Malaria. Infect Immun 2018; 86:IAI.00929-17. [PMID: 29784863 DOI: 10.1128/iai.00929-17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 05/18/2018] [Indexed: 02/07/2023] Open
Abstract
In animal models of experimental cerebral malaria (ECM), the glycosylphosphatidylinositols (GPIs) and GPI anchors are the major factors that induce nuclear factor kappa B (NF-κB) activation and proinflammatory responses, which contribute to malaria pathogenesis. GPIs and GPI anchors are transported to the cell surface via a process called GPI transamidation, which involves the GPI transamidase (GPI-T) complex. In this study, we showed that GPI16, one of the GPI-T subunits, is highly conserved among Plasmodium species. Genetic knockout of pbgpi16 (Δpbgpi16) in the rodent malaria parasite Plasmodium berghei strain ANKA led to a significant reduction of the amounts of GPIs in the membranes of merozoites, as well as surface display of several GPI-anchored merozoite surface proteins. Compared with the wild-type parasites, Δpbgpi16 parasites in C57BL/6 mice caused much less NF-κB activation and elicited a substantially attenuated T helper type 1 response. As a result, Δpbgpi16 mutant-infected mice displayed much less severe brain pathology, and considerably fewer Δpbgpi16 mutant-infected mice died from ECM. This study corroborated the GPI toxin as a significant inducer of ECM and further suggested that vaccines against parasite GPIs may be a promising strategy to limit the severity of malaria.
Collapse
|
25
|
Douglas RG, Nandekar P, Aktories JE, Kumar H, Weber R, Sattler JM, Singer M, Lepper S, Sadiq SK, Wade RC, Frischknecht F. Inter-subunit interactions drive divergent dynamics in mammalian and Plasmodium actin filaments. PLoS Biol 2018; 16:e2005345. [PMID: 30011270 PMCID: PMC6055528 DOI: 10.1371/journal.pbio.2005345] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 06/13/2018] [Indexed: 01/01/2023] Open
Abstract
Cell motility is essential for protozoan and metazoan organisms and typically relies on the dynamic turnover of actin filaments. In metazoans, monomeric actin polymerises into usually long and stable filaments, while some protozoans form only short and highly dynamic actin filaments. These different dynamics are partly due to the different sets of actin regulatory proteins and partly due to the sequence of actin itself. Here we probe the interactions of actin subunits within divergent actin filaments using a comparative dynamic molecular model and explore their functions using Plasmodium, the protozoan causing malaria, and mouse melanoma derived B16-F1 cells as model systems. Parasite actin tagged to a fluorescent protein (FP) did not incorporate into mammalian actin filaments, and rabbit actin-FP did not incorporate into parasite actin filaments. However, exchanging the most divergent region of actin subdomain 3 allowed such reciprocal incorporation. The exchange of a single amino acid residue in subdomain 2 (N41H) of Plasmodium actin markedly improved incorporation into mammalian filaments. In the parasite, modification of most subunit–subunit interaction sites was lethal, whereas changes in actin subdomains 1 and 4 reduced efficient parasite motility and hence mosquito organ penetration. The strong penetration defects could be rescued by overexpression of the actin filament regulator coronin. Through these comparative approaches we identified an essential and common contributor, subdomain 3, which drives the differential dynamic behaviour of two highly divergent eukaryotic actins in motile cells. Actin is one of the most abundant and conserved proteins across eukaryotes. Its ability to assemble from individual monomers into dynamic polymers is essential for many cellular functions, including division and motility. In most cells, actin is able to form long and stable filaments. However, an actin of the malaria-causing parasite Plasmodium, while having a very similar monomer structure to actins from other eukaryotes, forms only short and unstable filaments. These short and dynamic filaments are crucial in allowing the parasite to move very rapidly in tissue. Here we investigated the basis of these differences. We used molecular dynamics simulations of actin filaments to investigate the actin–actin interfaces in filaments from Plasmodium and rabbit. We next engineered parasites to express chimeric actins that contained different parts of rabbit and parasite actin and thereby identified actin residues important for parasite viability and progression across the life cycle. We could rescue the most prominent defect specifically with overexpression of the actin binding protein coronin. This suggests that the more stable actin harms the parasite and that coronin helps in recycling filaments. By screening the effects of actin chimeras in mammalian cells, we also identified regions that allow these different actins to efficiently interact with each other. Taken together, our results improve our understanding of the interactions required for actin to incorporate into filaments across divergent eukaryotes.
Collapse
Affiliation(s)
- Ross G. Douglas
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Prajwal Nandekar
- Molecular and Cellular Modeling, Heidelberg Institute for Theoretical Studies (HITS), Heidelberg, Germany
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Julia-Elisabeth Aktories
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Hirdesh Kumar
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
- Molecular and Cellular Modeling, Heidelberg Institute for Theoretical Studies (HITS), Heidelberg, Germany
| | - Rebekka Weber
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Julia M. Sattler
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Mirko Singer
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Simone Lepper
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - S. Kashif Sadiq
- Molecular and Cellular Modeling, Heidelberg Institute for Theoretical Studies (HITS), Heidelberg, Germany
| | - Rebecca C. Wade
- Molecular and Cellular Modeling, Heidelberg Institute for Theoretical Studies (HITS), Heidelberg, Germany
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
- Interdisciplinary Center for Scientific Computing (IWR), Heidelberg, Germany
- * E-mail: (FF); (RCW)
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
- * E-mail: (FF); (RCW)
| |
Collapse
|
26
|
Soga A, Ko‐ketsu M, Fukumoto S. Development of a
bsd
‐blasticidin selection system in
Plasmodium berghei. FEBS Lett 2018; 592:1847-1855. [DOI: 10.1002/1873-3468.13100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 11/06/2022]
Affiliation(s)
- Akira Soga
- National Research Center for Protozoan Diseases Obihiro University of Agriculture and Veterinary Medicine Japan
| | - Mami Ko‐ketsu
- National Research Center for Protozoan Diseases Obihiro University of Agriculture and Veterinary Medicine Japan
| | - Shinya Fukumoto
- National Research Center for Protozoan Diseases Obihiro University of Agriculture and Veterinary Medicine Japan
| |
Collapse
|
27
|
Böhme U, Otto TD, Cotton JA, Steinbiss S, Sanders M, Oyola SO, Nicot A, Gandon S, Patra KP, Herd C, Bushell E, Modrzynska KK, Billker O, Vinetz JM, Rivero A, Newbold CI, Berriman M. Complete avian malaria parasite genomes reveal features associated with lineage-specific evolution in birds and mammals. Genome Res 2018; 28:547-560. [PMID: 29500236 PMCID: PMC5880244 DOI: 10.1101/gr.218123.116] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/28/2018] [Indexed: 01/08/2023]
Abstract
Avian malaria parasites are prevalent around the world and infect a wide diversity of bird species. Here, we report the sequencing and analysis of high-quality draft genome sequences for two avian malaria species, Plasmodium relictum and Plasmodium gallinaceum We identify 50 genes that are specific to avian malaria, located in an otherwise conserved core of the genome that shares gene synteny with all other sequenced malaria genomes. Phylogenetic analysis suggests that the avian malaria species form an outgroup to the mammalian Plasmodium species, and using amino acid divergence between species, we estimate the avian- and mammalian-infective lineages diverged in the order of 10 million years ago. Consistent with their phylogenetic position, we identify orthologs of genes that had previously appeared to be restricted to the clades of parasites containing Plasmodium falciparum and Plasmodium vivax, the species with the greatest impact on human health. From these orthologs, we explore differential diversifying selection across the genus and show that the avian lineage is remarkable in the extent to which invasion-related genes are evolving. The subtelomeres of the P. relictum and P. gallinaceum genomes contain several novel gene families, including an expanded surf multigene family. We also identify an expansion of reticulocyte binding protein homologs in P. relictum, and within these proteins, we detect distinct regions that are specific to nonhuman primate, humans, rodent, and avian hosts. For the first time in the Plasmodium lineage, we find evidence of transposable elements, including several hundred fragments of LTR-retrotransposons in both species and an apparently complete LTR-retrotransposon in the genome of P. gallinaceum.
Collapse
Affiliation(s)
- Ulrike Böhme
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - Thomas D Otto
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - James A Cotton
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - Sascha Steinbiss
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - Mandy Sanders
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - Samuel O Oyola
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
- International Livestock Research Institute, Nairobi 00100, Kenya
| | - Antoine Nicot
- CEFE UMR 5175, CNRS-Université de Montpellier-Université Paul-Valéry Montpellier-EPHE, 34293 Montpellier Cedex 5, France
| | - Sylvain Gandon
- CEFE UMR 5175, CNRS-Université de Montpellier-Université Paul-Valéry Montpellier-EPHE, 34293 Montpellier Cedex 5, France
| | - Kailash P Patra
- Department of Medicine, Division of Infectious Diseases, University of California San Diego, School of Medicine, La Jolla, California 92093, USA
| | - Colin Herd
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - Ellen Bushell
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - Katarzyna K Modrzynska
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - Oliver Billker
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - Joseph M Vinetz
- Department of Medicine, Division of Infectious Diseases, University of California San Diego, School of Medicine, La Jolla, California 92093, USA
| | - Ana Rivero
- MIVEGEC (CNRS UMR 5290), 34394 Montpellier Cedex 5, France
| | - Chris I Newbold
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
- Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, United Kingdom
| | - Matthew Berriman
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, United Kingdom
| |
Collapse
|
28
|
Haeberlein S, Chevalley-Maurel S, Ozir-Fazalalikhan A, Koppejan H, Winkel BMF, Ramesar J, Khan SM, Sauerwein RW, Roestenberg M, Janse CJ, Smits HH, Franke-Fayard B. Protective immunity differs between routes of administration of attenuated malaria parasites independent of parasite liver load. Sci Rep 2017; 7:10372. [PMID: 28871201 PMCID: PMC5583236 DOI: 10.1038/s41598-017-10480-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 08/10/2017] [Indexed: 12/27/2022] Open
Abstract
In humans and murine models of malaria, intradermal immunization (ID-I) with genetically attenuated sporozoites that arrest in liver induces lower protective immunity than intravenous immunization (IV-I). It is unclear whether this difference is caused by fewer sporozoites migrating into the liver or by suboptimal hepatic and injection site-dependent immune responses. We therefore developed a Plasmodium yoelii immunization/boost/challenge model to examine parasite liver loads as well as hepatic and lymph node immune responses in protected and unprotected ID-I and IV-I animals. Despite introducing the same numbers of genetically attenuated parasites in the liver, ID-I resulted in lower sterile protection (53-68%) than IV-I (93-95%). Unprotected mice developed less sporozoite-specific CD8+ and CD4+ effector T-cell responses than protected mice. After immunization, ID-I mice showed more interleukin-10-producing B and T cells in livers and skin-draining lymph nodes, but fewer hepatic CD8 memory T cells and CD8+ dendritic cells compared to IV-I mice. Our results indicate that the lower protection efficacy obtained by intradermal sporozoite administration is not linked to low hepatic parasite numbers as presumed before, but correlates with a shift towards regulatory immune responses. Overcoming these immune suppressive responses is important not only for live-attenuated malaria vaccines but also for other live vaccines administered in the skin.
Collapse
Affiliation(s)
- Simone Haeberlein
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.,Institute of Parasitology, Justus-Liebig-University Giessen, Schubertstrasse 81, 35392, Giessen, Germany
| | - Séverine Chevalley-Maurel
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Arifa Ozir-Fazalalikhan
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Hester Koppejan
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Beatrice M F Winkel
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Jai Ramesar
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Shahid M Khan
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, Geert-Grooteplein 28, 6525 GA, Nijmegen, The Netherlands
| | - Meta Roestenberg
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.,Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Chris J Janse
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Hermelijn H Smits
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Blandine Franke-Fayard
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
29
|
Plasmodium berghei PIMMS2 Promotes Ookinete Invasion of the Anopheles gambiae Mosquito Midgut. Infect Immun 2017; 85:IAI.00139-17. [PMID: 28559405 PMCID: PMC5520436 DOI: 10.1128/iai.00139-17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 05/11/2017] [Indexed: 12/21/2022] Open
Abstract
Mosquito midgut stages of the malaria parasite present an attractive biological system to study host-parasite interactions and develop interventions to block disease transmission. Mosquito infection ensues upon oocyst development that follows ookinete invasion and traversal of the mosquito midgut epithelium. Here, we report the characterization of PIMMS2 (Plasmodium invasion of mosquito midgut screen candidate 2), a Plasmodium berghei protein with structural similarities to subtilisin-like proteins. PIMMS2 orthologs are present in the genomes of all plasmodia and are mapped between the subtilisin-encoding genes SUB1 and SUB3. P. berghei PIMMS2 is specifically expressed in zygotes and ookinetes and is localized on the ookinete surface. Loss of PIMMS2 function through gene disruption by homologous recombination leads to normal development of motile ookinetes that exhibit a severely impaired capacity to traverse the mosquito midgut and transform to oocysts. Genetic complementation of the disrupted locus with a mutated PIMMS2 allele reveals that amino acid residues corresponding to the putative subtilisin-like catalytic triad are important but not essential for protein function. Our data demonstrate that PIMMS2 is a novel ookinete-specific protein that promotes parasite traversal of the mosquito midgut epithelium and establishment of mosquito infection.
Collapse
|
30
|
Soga A, Bando H, Ko-Ketsu M, Masuda-Suganuma H, Kawazu SI, Fukumoto S. High efficacy in vitro selection procedure for generating transgenic parasites of Plasmodium berghei using an antibiotic toxic to rodent hosts. Sci Rep 2017. [PMID: 28638105 PMCID: PMC5479828 DOI: 10.1038/s41598-017-04244-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The malaria parasite Plasmodium berghei is one of the main rodent malaria models. A shortcoming of this model parasite is its low flexibility in genetic manipulation. As this parasite cannot be continuously propagated in cell cultures, in vivo drug selection procedures are necessary to isolate genetic mutants. Drugs harmful to rodents therefore cannot be used for drug selection, which restricts the range of genetic manipulation. In this study, we addressed this problem by establishing a novel in vitro culture drug selection method, which we used in combination with other established methods to successfully isolate genetically manipulated parasites. The target mutants were enriched to the desired level within two weeks. We show that our system can also be used for sequential genetic manipulation of parasites carrying the traditionally used selection markers, demonstrate the procedure’s versatility, and show its use in isolating specific genetically manipulated parasites. This novel in vitro selection method increases the number of available selection markers, allowing more extensive genetic manipulation in malaria parasite research.
Collapse
Affiliation(s)
- Akira Soga
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan
| | - Hironori Bando
- Department of immunoparasitology, Research Institute for Microbial Disease, Osaka University, Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Mami Ko-Ketsu
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan
| | - Hirono Masuda-Suganuma
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan
| | - Shin-Ichiro Kawazu
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan
| | - Shinya Fukumoto
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido, 080-8555, Japan.
| |
Collapse
|
31
|
Klug D, Frischknecht F. Motility precedes egress of malaria parasites from oocysts. eLife 2017; 6. [PMID: 28115054 PMCID: PMC5262382 DOI: 10.7554/elife.19157] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 12/16/2016] [Indexed: 12/19/2022] Open
Abstract
Malaria is transmitted when an infected Anopheles mosquito deposits Plasmodium sporozoites in the skin during a bite. Sporozoites are formed within oocysts at the mosquito midgut wall and are released into the hemolymph, from where they invade the salivary glands and are subsequently transmitted to the vertebrate host. We found that a thrombospondin-repeat containing sporozoite-specific protein named thrombospondin-releated protein 1 (TRP1) is important for oocyst egress and salivary gland invasion, and hence for the transmission of malaria. We imaged the release of sporozoites from oocysts in situ, which was preceded by active motility. Parasites lacking TRP1 failed to migrate within oocysts and did not egress, suggesting that TRP1 is a vital component of the events that precede intra-oocyst motility and subsequently sporozoite egress and salivary gland invasion. DOI:http://dx.doi.org/10.7554/eLife.19157.001 Malaria is caused by a parasite transmitted by certain types of mosquito. The parasite lives in different organs within its vertebrate animal and insect hosts and to cope with these different environments it has a complex life cycle with several highly specialized life stages. To move from an infected mosquito into vertebrates the parasite produces spore-like cells called sporozoites that are able to enter different tissues and move very fast. These cells develop inside parasite-made structures called oocysts, which form at the stomach wall of the mosquito. After emerging from the oocyst, sporozoites float through the mosquito’s circulatory system and eventually enter the salivary glands where they can be transmitted to vertebrates when the mosquito bites. Efforts to develop malaria treatments and vaccines have focused on understanding the parasite’s life cycle and identifying ways to control or eradicate key stages. Most researchers focus on the stage where the parasite is living in the vertebrate and actively causing disease, while the events in the mosquito are less intensely investigated. While several parasite proteins have been shown to be important for the release of sporozoites from oocysts, the molecular events leading to this release have not yet been fully resolved. Klug and Frischknecht used time-lapse microscopy to film the release of the sporozoites of a malaria parasite known as Plasmodium berghei. The experiments show that the sporozoites can leave oocysts in several different ways. Furthermore, Klug and Frischknecht identified a new parasite protein named TRP1 that is essential for the sporozoites to leave oocysts and invade the salivary glands. Sporozoites lacking TRP1 were not able to move and they were unable to leave the oocyst or invade the salivary glands. Klug and Frischknecht propose a new working model of the molecular events that govern sporozoite release in which TRP1 is required for sporozoites to move prior to their exit from oocysts. In the future, using the same techniques to analyze genetically modified parasites will help to reveal more details about sporozoite release. DOI:http://dx.doi.org/10.7554/eLife.19157.002
Collapse
Affiliation(s)
- Dennis Klug
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| |
Collapse
|
32
|
Klug D, Mair GR, Frischknecht F, Douglas RG. A small mitochondrial protein present in myzozoans is essential for malaria transmission. Open Biol 2016; 6:160034. [PMID: 27053680 PMCID: PMC4852462 DOI: 10.1098/rsob.160034] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Myzozoans (which include dinoflagellates, chromerids and apicomplexans) display notable divergence from their ciliate sister group, including a reduced mitochondrial genome and divergent metabolic processes. The factors contributing to these divergent processes are still poorly understood and could serve as potential drug targets in disease-causing protists. Here, we report the identification and characterization of a small mitochondrial protein from the rodent-infecting apicomplexan parasite Plasmodium berghei that is essential for development in its mosquito host. Parasites lacking the gene mitochondrial protein ookinete developmental defect (mpodd) showed malformed parasites that were unable to transmit to mosquitoes. Knockout parasites displayed reduced mitochondrial mass without affecting organelle integrity, indicating no role of the protein in mitochondrial biogenesis or morphology maintenance but a likely role in mitochondrial import or metabolism. Using genetic complementation experiments, we identified a previously unrecognized Plasmodium falciparum homologue that can rescue the mpodd(−) phenotype, thereby showing that the gene is functionally conserved. As far as can be detected, mpodd is found in myzozoans, has homologues in the phylum Apicomplexa and appears to have arisen in free-living dinoflagellates. This suggests that the MPODD protein has a conserved mitochondrial role that is important for myzozoans. While previous studies identified a number of essential proteins which are generally highly conserved evolutionarily, our study identifies, for the first time, a non-canonical protein fulfilling a crucial function in the mitochondrion during parasite transmission.
Collapse
Affiliation(s)
- Dennis Klug
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Gunnar R Mair
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Ross G Douglas
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| |
Collapse
|
33
|
Rapid Generation of Marker-Free P. falciparum Fluorescent Reporter Lines Using Modified CRISPR/Cas9 Constructs and Selection Protocol. PLoS One 2016; 11:e0168362. [PMID: 27997583 PMCID: PMC5172577 DOI: 10.1371/journal.pone.0168362] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 11/30/2016] [Indexed: 01/19/2023] Open
Abstract
The CRISPR/Cas9 system is a powerful genome editing technique employed in a wide variety of organisms including recently the human malaria parasite, P. falciparum. Here we report on further improvements to the CRISPR/Cas9 transfection constructs and selection protocol to more rapidly modify the P. falciparum genome and to introduce transgenes into the parasite genome without the inclusion of drug-selectable marker genes. This method was used to stably integrate the gene encoding GFP into the P. falciparum genome under the control of promoters of three different Plasmodium genes (calmodulin, gapdh and hsp70). These genes were selected as they are highly transcribed in blood stages. We show that the three reporter parasite lines generated in this study (GFP@cam, GFP@gapdh and GFP@hsp70) have in vitro blood stage growth kinetics and drug-sensitivity profiles comparable to the parental P. falciparum (NF54) wild-type line. Both asexual and sexual blood stages of the three reporter lines expressed GFP-fluorescence with GFP@hsp70 having the highest fluorescent intensity in schizont stages as shown by flow cytometry analysis of GFP-fluorescence intensity. The improved CRISPR/Cas9 constructs/protocol will aid in the rapid generation of transgenic and modified P. falciparum parasites, including those expressing different reporters proteins under different (stage specific) promoters.
Collapse
|
34
|
Fougère A, Jackson AP, Paraskevi Bechtsi D, Braks JAM, Annoura T, Fonager J, Spaccapelo R, Ramesar J, Chevalley-Maurel S, Klop O, van der Laan AMA, Tanke HJ, Kocken CHM, Pasini EM, Khan SM, Böhme U, van Ooij C, Otto TD, Janse CJ, Franke-Fayard B. Variant Exported Blood-Stage Proteins Encoded by Plasmodium Multigene Families Are Expressed in Liver Stages Where They Are Exported into the Parasitophorous Vacuole. PLoS Pathog 2016; 12:e1005917. [PMID: 27851824 PMCID: PMC5113031 DOI: 10.1371/journal.ppat.1005917] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 09/06/2016] [Indexed: 01/05/2023] Open
Abstract
Many variant proteins encoded by Plasmodium-specific multigene families are exported into red blood cells (RBC). P. falciparum-specific variant proteins encoded by the var, stevor and rifin multigene families are exported onto the surface of infected red blood cells (iRBC) and mediate interactions between iRBC and host cells resulting in tissue sequestration and rosetting. However, the precise function of most other Plasmodium multigene families encoding exported proteins is unknown. To understand the role of RBC-exported proteins of rodent malaria parasites (RMP) we analysed the expression and cellular location by fluorescent-tagging of members of the pir, fam-a and fam-b multigene families. Furthermore, we performed phylogenetic analyses of the fam-a and fam-b multigene families, which indicate that both families have a history of functional differentiation unique to RMP. We demonstrate for all three families that expression of family members in iRBC is not mutually exclusive. Most tagged proteins were transported into the iRBC cytoplasm but not onto the iRBC plasma membrane, indicating that they are unlikely to play a direct role in iRBC-host cell interactions. Unexpectedly, most family members are also expressed during the liver stage, where they are transported into the parasitophorous vacuole. This suggests that these protein families promote parasite development in both the liver and blood, either by supporting parasite development within hepatocytes and erythrocytes and/or by manipulating the host immune response. Indeed, in the case of Fam-A, which have a steroidogenic acute regulatory-related lipid transfer (START) domain, we found that several family members can transfer phosphatidylcholine in vitro. These observations indicate that these proteins may transport (host) phosphatidylcholine for membrane synthesis. This is the first demonstration of a biological function of any exported variant protein family of rodent malaria parasites. Malaria-parasites invade and multiply in hepatocytes and erythrocytes. The human parasite P. falciparum transports proteins encoded by multigene families onto the surface of erythrocytes, mediating interactions between infected red blood cells (iRBCs) and other host-cells and are thought to play a key role in parasite survival during blood-stage development. The function of other exported Plasmodium protein families remains largely unknown. We provide novel insights into expression and cellular location of proteins encoded by three large multigene families of rodent malaria parasites (Fam-a, Fam-b and PIR). Multiple members of the same family are expressed in a single iRBC, unlike P. falciparum PfEMP1 proteins where individual iRBCs express only a single member. Most proteins we examined are located in the RBC cytoplasm and are not transported onto the iRBC surface membrane, indicating that these proteins are unlikely to mediate interactions between iRBCs and host-cells. Unexpectedly, liver stages also express many of these proteins, where they locate to the vacuole surrounding the parasite inside the hepatocyte. In support of a role of these proteins for parasite growth within their host cells we provide evidence that Fam-A proteins have a role in uptake and transport of (host) phosphatidylcholine for parasite-membrane synthesis.
Collapse
Affiliation(s)
- Aurélie Fougère
- Leiden Malaria Research Group, Parasitology, Center of infectious Diseases, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Department of Experimental Medicine, University of Perugia, Italy
| | - Andrew P. Jackson
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool, UnitedKingdom
| | | | - Joanna A. M. Braks
- Leiden Malaria Research Group, Parasitology, Center of infectious Diseases, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Takeshi Annoura
- Leiden Malaria Research Group, Parasitology, Center of infectious Diseases, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Department of Department of Parasitology, National Institute of Infectious Diseases (NIID), Tokyo, Japan
| | - Jannik Fonager
- Leiden Malaria Research Group, Parasitology, Center of infectious Diseases, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Department of Microbiological Diagnostics and Virology, Statens Serum Institute, Copenhagen, Denmark
| | | | - Jai Ramesar
- Leiden Malaria Research Group, Parasitology, Center of infectious Diseases, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Séverine Chevalley-Maurel
- Leiden Malaria Research Group, Parasitology, Center of infectious Diseases, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Onny Klop
- Leiden Malaria Research Group, Parasitology, Center of infectious Diseases, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | | | - Hans J. Tanke
- Department of Molecular Cell Biology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | | | - Erica M. Pasini
- Biomedical Primate Research Centre (BPRC), Rijswijk, The Netherlands
| | - Shahid M. Khan
- Leiden Malaria Research Group, Parasitology, Center of infectious Diseases, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Ulrike Böhme
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UnitedKingdom
| | - Christiaan van Ooij
- The Francis Crick Institute, Mill Hill Laboratory, Mill Hill, London, UnitedKingdom
| | - Thomas D. Otto
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UnitedKingdom
| | - Chris J. Janse
- Leiden Malaria Research Group, Parasitology, Center of infectious Diseases, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Blandine Franke-Fayard
- Leiden Malaria Research Group, Parasitology, Center of infectious Diseases, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
35
|
Posayapisit N, Songsungthong W, Koonyosying P, Falade MO, Uthaipibull C, Yuthavong Y, Shaw PJ, Kamchonwongpaisan S. Cytochrome c and c1 heme lyases are essential in Plasmodium berghei. Mol Biochem Parasitol 2016; 210:32-36. [PMID: 27520480 DOI: 10.1016/j.molbiopara.2016.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 08/04/2016] [Accepted: 08/08/2016] [Indexed: 10/21/2022]
Abstract
Malaria parasites possess a de novo heme synthetic pathway. Interestingly, this pathway is dispensable during the blood stages of development in mammalian hosts. The assembly of the two most important hemeproteins, cytochromes c and c1, is mediated by cytochrome heme lyase enzymes. Plasmodium spp. possess two cytochrome heme lyases encoded by separate genes. Given the redundancy of heme synthesis, we sought to determine if heme lyase function also exhibits redundancy. To answer this question, we performed gene knockout experiments. We found that the PBANKA_143950 and PBANKA_0602600 Plasmodium berghei genes encoding cytochrome c (Pbcchl) and cytochrome c1 (Pbcc1hl) heme lyases, respectively, can only be disrupted when a complementary gene is present. In contrast, four genes in the de novo heme synthesis pathway can be disrupted without complementation. This work provides evidence that Pbcchl and Pbcc1hl are both essential and thus may be antimalarial targets.
Collapse
Affiliation(s)
- Navaporn Posayapisit
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Thanon Phahonyothin, Tambon Khlong Neung, Amphoe Khlong Luang, Pathum Thani 12120, Thailand
| | - Warangkhana Songsungthong
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Thanon Phahonyothin, Tambon Khlong Neung, Amphoe Khlong Luang, Pathum Thani 12120, Thailand
| | - Pongpisid Koonyosying
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Thanon Phahonyothin, Tambon Khlong Neung, Amphoe Khlong Luang, Pathum Thani 12120, Thailand
| | - Mofolusho O Falade
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Thanon Phahonyothin, Tambon Khlong Neung, Amphoe Khlong Luang, Pathum Thani 12120, Thailand
| | - Chairat Uthaipibull
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Thanon Phahonyothin, Tambon Khlong Neung, Amphoe Khlong Luang, Pathum Thani 12120, Thailand
| | - Yongyuth Yuthavong
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Thanon Phahonyothin, Tambon Khlong Neung, Amphoe Khlong Luang, Pathum Thani 12120, Thailand
| | - Philip J Shaw
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Thanon Phahonyothin, Tambon Khlong Neung, Amphoe Khlong Luang, Pathum Thani 12120, Thailand.
| | - Sumalee Kamchonwongpaisan
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Thanon Phahonyothin, Tambon Khlong Neung, Amphoe Khlong Luang, Pathum Thani 12120, Thailand.
| |
Collapse
|
36
|
Pornthanakasem W, Riangrungroj P, Chitnumsub P, Ittarat W, Kongkasuriyachai D, Uthaipibull C, Yuthavong Y, Leartsakulpanich U. Role of Plasmodium vivax Dihydropteroate Synthase Polymorphisms in Sulfa Drug Resistance. Antimicrob Agents Chemother 2016; 60:4453-63. [PMID: 27161627 PMCID: PMC4958149 DOI: 10.1128/aac.01835-15] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 04/19/2016] [Indexed: 11/20/2022] Open
Abstract
Dihydropteroate synthase (DHPS) is a known sulfa drug target in malaria treatment, existing as a bifunctional enzyme together with hydroxymethyldihydropterin pyrophosphokinase (HPPK). Polymorphisms in key residues of Plasmodium falciparum DHPS (PfDHPS) have been characterized and linked to sulfa drug resistance in malaria. Genetic sequencing of P. vivax dhps (Pvdhps) from clinical isolates has shown several polymorphisms at the positions equivalent to those in the Pfdhps genes conferring sulfa drug resistance, suggesting a mechanism for sulfa drug resistance in P. vivax similar to that seen in P. falciparum To characterize the role of polymorphisms in the PvDHPS in sulfa drug resistance, various mutants of recombinant PvHPPK-DHPS enzymes were expressed and characterized. Moreover, due to the lack of a continuous in vitro culture system for P. vivax parasites, a surrogate P. berghei model expressing Pvhppk-dhps genes was established to demonstrate the relationship between sequence polymorphisms and sulfa drug susceptibility and to test the activities of PvDHPS inhibitors on the transgenic parasites. Both enzyme activity and transgenic parasite growth were sensitive to sulfadoxine to different degrees, depending on the number of mutations that accumulated in DHPS. Ki values and 50% effective doses were higher for mutant PvDHPS enzymes than the wild-type enzymes. Altogether, the study provides the first evidence of sulfa drug resistance at the molecular level in P. vivax Furthermore, the enzyme inhibition assay and the in vivo screening system can be useful tools for screening new compounds for their activities against PvDHPS.
Collapse
Affiliation(s)
| | | | - Penchit Chitnumsub
- National Center for Genetic Engineering and Biotechnology, Pathum Thani, Thailand
| | - Wanwipa Ittarat
- National Center for Genetic Engineering and Biotechnology, Pathum Thani, Thailand
| | | | - Chairat Uthaipibull
- National Center for Genetic Engineering and Biotechnology, Pathum Thani, Thailand
| | - Yongyuth Yuthavong
- National Center for Genetic Engineering and Biotechnology, Pathum Thani, Thailand
| | | |
Collapse
|
37
|
Kou X, Zheng W, Du F, Liu F, Wang M, Fan Q, Cui L, Luo E, Cao Y. Characterization of a Plasmodium berghei sexual stage antigen PbPH as a new candidate for malaria transmission-blocking vaccine. Parasit Vectors 2016; 9:190. [PMID: 27038925 PMCID: PMC4818878 DOI: 10.1186/s13071-016-1459-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 03/15/2016] [Indexed: 12/01/2022] Open
Abstract
Background Transmission-blocking vaccines (TBVs) are a promising strategy for malaria control and elimination. However, candidate TBV antigens are currently limited, highlighting the urgency of identifying new antigens for TBV development. Methods Using a combination of bioinformatic analysis and functional studies in the rodent malaria model Plasmodium berghei, we identified a conserved Plasmodium protein PbPH (PBANKA_041720) containing a pleckstrin homology (PH) domain. The expression of PbPH was detected by Western blot and indirect immunofluorescence assay (IFA). The function of PbPH was tested by genetic knockout. The TB activity was confirmed by in vitro ookinete conversion assay and mosquito feeding. Results PbPH was detected in Western blot as highly expressed in sexual stages (gametocytes and ookinetes). IFA revealed localizations of PbPH on the surface of gametes, zygotes, and ookinetes. Deletion of the pbph gene did not affect asexual growth, but significantly reduced the formation of gametocytes, ookinetes, and oocysts, indicating that PbPH protein is required for parasite sexual development. Recombinant PbPH expressed and purified from bacteria elicited strong antibody responses in mice and the antibodies significantly inhibited exflagellation of male gametocytes and formation of ookinetes in a concentration-dependent manner. Mosquito feeding experiments confirmed that mosquitoes fed on mice immunized with PbPH had 13 % reduction in the prevalence of infection and almost 48 % reduction in oocyst density. Conclusions Pbph is a highly conserved Plasmodium gene and is required for parasite sexual development. PbPH protein is expressed on the surface of gametes and ookinetes. Immunization of mice against the recombinant PbPH protein induced strong antibody responses that effectively reduced the formation of male gametes and ookinetes in vitro and blocked transmission of the parasites to mosquitoes. These results highlight PbPH as a potential TBV candidate that is worth future investigations in human malaria parasites. Electronic supplementary material The online version of this article (doi:10.1186/s13071-016-1459-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xu Kou
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110001, China.,College of Animal Husbandry and Veterinary, Liaoning Medical University, Jinzhou, Liaoning, 121001, China
| | - Wenqi Zheng
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110001, China
| | - Feng Du
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110001, China
| | - Fei Liu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110001, China
| | - Meilian Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110001, China
| | - Qi Fan
- Dalian Institute of Biotechnology, Dalian, Liaoning, China
| | - Liwang Cui
- Department of Entomology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Enjie Luo
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110001, China.
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
38
|
Singer M, Marshall J, Heiss K, Mair GR, Grimm D, Mueller AK, Frischknecht F. Zinc finger nuclease-based double-strand breaks attenuate malaria parasites and reveal rare microhomology-mediated end joining. Genome Biol 2015; 16:249. [PMID: 26573820 PMCID: PMC4647826 DOI: 10.1186/s13059-015-0811-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/22/2015] [Indexed: 02/06/2023] Open
Abstract
Background Genome editing of malaria parasites is key to the generation of live attenuated parasites used in experimental vaccination approaches. DNA repair in Plasmodium generally occurs only through homologous recombination. This has been used to generate transgenic parasites that lack one to three genes, leading to developmental arrest in the liver and allowing the host to launch a protective immune response. While effective in principle, this approach is not safe for use in humans as single surviving parasites can still cause disease. Here we use zinc-finger nucleases to generate attenuated parasite lines lacking an entire chromosome arm, by a timed induction of a double-strand break. Rare surviving parasites also allow the investigation of unconventional DNA repair mechanisms in a rodent malaria parasite. Results A single, zinc-finger nuclease-induced DNA double-strand break results in the generation of attenuated parasite lines that show varying degrees of developmental arrest, protection efficacy in an immunisation regime and safety, depending on the timing of zinc-finger nuclease expression within the life cycle. We also identify DNA repair by microhomology-mediated end joining with as little as four base pairs, resulting in surviving parasites and thus breakthrough infections. Conclusions Malaria parasites can repair DNA double-strand breaks with surprisingly small mini-homology domains located across the break point. Timely expression of zinc-finger nucleases could be used to generate a new generation of attenuated parasite lines lacking hundreds of genes. Electronic supplementary material The online version of this article (doi:10.1186/s13059-015-0811-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mirko Singer
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany.
| | - Jennifer Marshall
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Kirsten Heiss
- Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany.,MalVa GmbH, Heidelberg, Germany
| | - Gunnar R Mair
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Dirk Grimm
- Virology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany
| | - Ann-Kristin Mueller
- Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany.,German Center for Infectious Diseases, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany.
| |
Collapse
|
39
|
Shao M, Michno JM, Hotton SK, Blechl A, Thomson J. A bacterial gene codA encoding cytosine deaminase is an effective conditional negative selectable marker in Glycine max. PLANT CELL REPORTS 2015; 34:1707-16. [PMID: 26082433 DOI: 10.1007/s00299-015-1818-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 05/22/2015] [Accepted: 06/01/2015] [Indexed: 06/04/2023]
Abstract
KEY MESSAGE Research describes the practical application of the codA negative selection marker in Soybean. Conditions are given for codA selection at both the shooting and rooting stages of regeneration. Conditional negative selection is a powerful technique whereby the absence of a gene product allows survival in otherwise lethal conditions. In plants, the Escherichia coli gene codA has been employed as a negative selection marker. Our research demonstrates that codA can be used as a negative selection marker in soybean, Glycine max. Like most plants, soybean does not contain cytosine deaminase activity and we show here that wild-type seedlings are not affected by inclusion of 5-FC in growth media. In contrast, transgenic G. max plants expressing codA and grown in the presence of more than 200 μg/mL 5-FC exhibit reductions in hypocotyl and taproot lengths, and severe suppression of lateral root development. We also demonstrate a novel negative selection-rooting assay in which codA-expressing aerial tissues or shoot cuttings are inhibited for root formation in media containing 5-FC. Taken together these techniques allow screening during either the regeneration or rooting phase of tissue culture.
Collapse
Affiliation(s)
- Min Shao
- Department of Plant Sciences, University of California-Davis, Davis, CA, 95616, USA
| | - Jean-Michel Michno
- Department of Agronomy and Plant Genetics, University of Minnesota, St. Paul, MN, 55108, USA
| | - Sara K Hotton
- Department of Plant Sciences, University of California-Davis, Davis, CA, 95616, USA
| | - Ann Blechl
- Western Regional Research Center, USDA-ARS Crop Improvement and Genetics, Albany, CA, 94710, USA
| | - James Thomson
- Western Regional Research Center, USDA-ARS Crop Improvement and Genetics, Albany, CA, 94710, USA.
| |
Collapse
|
40
|
de Koning-Ward TF, Gilson PR, Crabb BS. Advances in molecular genetic systems in malaria. Nat Rev Microbiol 2015; 13:373-87. [DOI: 10.1038/nrmicro3450] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
41
|
Lin JW, Spaccapelo R, Schwarzer E, Sajid M, Annoura T, Deroost K, Ravelli RBG, Aime E, Capuccini B, Mommaas-Kienhuis AM, O'Toole T, Prins F, Franke-Fayard BMD, Ramesar J, Chevalley-Maurel S, Kroeze H, Koster AJ, Tanke HJ, Crisanti A, Langhorne J, Arese P, Van den Steen PE, Janse CJ, Khan SM. Replication of Plasmodium in reticulocytes can occur without hemozoin formation, resulting in chloroquine resistance. ACTA ACUST UNITED AC 2015; 212:893-903. [PMID: 25941254 PMCID: PMC4451122 DOI: 10.1084/jem.20141731] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 04/08/2015] [Indexed: 01/03/2023]
Abstract
Lin et al. generate Plasmodium berghei mutants lacking enzymes critical to hemoglobin digestion. A double gene deletion mutant lacking enzymes involved in the initial steps of hemoglobin proteolysis is able to replicate inside reticulocytes of infected mice with limited hemoglobin degradation and no hemozoin formation, and moreover, is resistant to the antimalarial drug chloroquine. Most studies on malaria-parasite digestion of hemoglobin (Hb) have been performed using P. falciparum maintained in mature erythrocytes, in vitro. In this study, we examine Plasmodium Hb degradation in vivo in mice, using the parasite P. berghei, and show that it is possible to create mutant parasites lacking enzymes involved in the initial steps of Hb proteolysis. These mutants only complete development in reticulocytes and mature into both schizonts and gametocytes. Hb degradation is severely impaired and large amounts of undigested Hb remains in the reticulocyte cytoplasm and in vesicles in the parasite. The mutants produce little or no hemozoin (Hz), the detoxification by-product of Hb degradation. Further, they are resistant to chloroquine, an antimalarial drug that interferes with Hz formation, but their sensitivity to artesunate, also thought to be dependent on Hb degradation, is retained. Survival in reticulocytes with reduced or absent Hb digestion may imply a novel mechanism of drug resistance. These findings have implications for drug development against human-malaria parasites, such as P. vivax and P. ovale, which develop inside reticulocytes.
Collapse
Affiliation(s)
- Jing-Wen Lin
- Leiden Malaria Research Group, Department of Parasitology, Department of Molecular Cell Biology, and Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands Division of Parasitology, MRC National Institute for Medical Research, London NW7 1AA, England, UK
| | - Roberta Spaccapelo
- Department of Experimental Medicine, University of Perugia, Piazzale Gambuli, 06132 Perugia, Italy
| | - Evelin Schwarzer
- Department of Oncology, University of Torino, 10124 Torino, Italy
| | - Mohammed Sajid
- Leiden Malaria Research Group, Department of Parasitology, Department of Molecular Cell Biology, and Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands
| | - Takeshi Annoura
- Leiden Malaria Research Group, Department of Parasitology, Department of Molecular Cell Biology, and Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands
| | - Katrien Deroost
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven - University of Leuven, 3000 Leuven, Belgium
| | - Raimond B G Ravelli
- Leiden Malaria Research Group, Department of Parasitology, Department of Molecular Cell Biology, and Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands
| | - Elena Aime
- Department of Experimental Medicine, University of Perugia, Piazzale Gambuli, 06132 Perugia, Italy
| | - Barbara Capuccini
- Department of Experimental Medicine, University of Perugia, Piazzale Gambuli, 06132 Perugia, Italy Division of Parasitology, MRC National Institute for Medical Research, London NW7 1AA, England, UK
| | - Anna M Mommaas-Kienhuis
- Leiden Malaria Research Group, Department of Parasitology, Department of Molecular Cell Biology, and Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands
| | - Tom O'Toole
- Department of Molecular Cell Biology and Immunology, Vrije University Medical Center, 1007 MB Amsterdam, Netherlands
| | - Frans Prins
- Leiden Malaria Research Group, Department of Parasitology, Department of Molecular Cell Biology, and Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands
| | - Blandine M D Franke-Fayard
- Leiden Malaria Research Group, Department of Parasitology, Department of Molecular Cell Biology, and Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands
| | - Jai Ramesar
- Leiden Malaria Research Group, Department of Parasitology, Department of Molecular Cell Biology, and Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands
| | - Séverine Chevalley-Maurel
- Leiden Malaria Research Group, Department of Parasitology, Department of Molecular Cell Biology, and Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands
| | - Hans Kroeze
- Leiden Malaria Research Group, Department of Parasitology, Department of Molecular Cell Biology, and Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands
| | - Abraham J Koster
- Leiden Malaria Research Group, Department of Parasitology, Department of Molecular Cell Biology, and Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands
| | - Hans J Tanke
- Leiden Malaria Research Group, Department of Parasitology, Department of Molecular Cell Biology, and Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands
| | - Andrea Crisanti
- Department of Experimental Medicine, University of Perugia, Piazzale Gambuli, 06132 Perugia, Italy Department of Biological Sciences, Imperial College London, South Kensington Campus, SAF, London SW7 2AZ, England, UK
| | - Jean Langhorne
- Division of Parasitology, MRC National Institute for Medical Research, London NW7 1AA, England, UK
| | - Paolo Arese
- Department of Oncology, University of Torino, 10124 Torino, Italy
| | - Philippe E Van den Steen
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven - University of Leuven, 3000 Leuven, Belgium
| | - Chris J Janse
- Leiden Malaria Research Group, Department of Parasitology, Department of Molecular Cell Biology, and Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands
| | - Shahid M Khan
- Leiden Malaria Research Group, Department of Parasitology, Department of Molecular Cell Biology, and Department of Pathology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands
| |
Collapse
|
42
|
Matz JM, Kooij TWA. Towards genome-wide experimental genetics in the in vivo malaria model parasite Plasmodium berghei. Pathog Glob Health 2015; 109:46-60. [PMID: 25789828 DOI: 10.1179/2047773215y.0000000006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Plasmodium berghei was identified as a parasite of thicket rats (Grammomys dolichurus) and Anopheles dureni mosquitoes in African highland forests. Successful adaptation to a range of rodent and mosquito species established P. berghei as a malaria model parasite. The introduction of stable transfection technology, permitted classical reverse genetics strategies and thus systematic functional profiling of the gene repertoire. In the past 10 years following the publication of the P. berghei genome sequence, many new tools for experimental genetics approaches have been developed and existing ones have been improved. The infection of mice is the principal limitation towards a genome-wide repository of mutant parasite lines. In the past few years, there have been some promising and most welcome developments that allow rapid selection and isolation of recombinant parasites while simultaneously minimising animal usage. Here, we provide an overview of all the currently available tools and methods.
Collapse
|
43
|
Gomes AR, Bushell E, Schwach F, Girling G, Anar B, Quail MA, Herd C, Pfander C, Modrzynska K, Rayner JC, Billker O. A genome-scale vector resource enables high-throughput reverse genetic screening in a malaria parasite. Cell Host Microbe 2015; 17:404-413. [PMID: 25732065 PMCID: PMC4362957 DOI: 10.1016/j.chom.2015.01.014] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 12/01/2014] [Accepted: 01/07/2015] [Indexed: 10/30/2022]
Abstract
The genome-wide identification of gene functions in malaria parasites is hampered by a lack of reverse genetic screening methods. We present a large-scale resource of barcoded vectors with long homology arms for effective modification of the Plasmodium berghei genome. Cotransfecting dozens of vectors into the haploid blood stages creates complex pools of barcoded mutants, whose competitive fitness can be measured during infection of a single mouse using barcode sequencing (barseq). To validate the utility of this resource, we rescreen the P. berghei kinome, using published kinome screens for comparison. We find that several protein kinases function redundantly in asexual blood stages and confirm the targetability of kinases cdpk1, gsk3, tkl3, and PBANKA_082960 by genotyping cloned mutants. Thus, parallel phenotyping of barcoded mutants unlocks the power of reverse genetic screening for a malaria parasite and will enable the systematic identification of genes essential for in vivo parasite growth and transmission.
Collapse
Affiliation(s)
- Ana Rita Gomes
- Wellcome Trust Sanger Institute, Hinxton Cambridge CB10 1SA, UK
| | - Ellen Bushell
- Wellcome Trust Sanger Institute, Hinxton Cambridge CB10 1SA, UK
| | - Frank Schwach
- Wellcome Trust Sanger Institute, Hinxton Cambridge CB10 1SA, UK
| | - Gareth Girling
- Wellcome Trust Sanger Institute, Hinxton Cambridge CB10 1SA, UK
| | - Burcu Anar
- Wellcome Trust Sanger Institute, Hinxton Cambridge CB10 1SA, UK
| | - Michael A Quail
- Wellcome Trust Sanger Institute, Hinxton Cambridge CB10 1SA, UK
| | - Colin Herd
- Wellcome Trust Sanger Institute, Hinxton Cambridge CB10 1SA, UK
| | - Claudia Pfander
- Wellcome Trust Sanger Institute, Hinxton Cambridge CB10 1SA, UK
| | | | - Julian C Rayner
- Wellcome Trust Sanger Institute, Hinxton Cambridge CB10 1SA, UK.
| | - Oliver Billker
- Wellcome Trust Sanger Institute, Hinxton Cambridge CB10 1SA, UK.
| |
Collapse
|
44
|
Manzoni G, Briquet S, Risco-Castillo V, Gaultier C, Topçu S, Ivănescu ML, Franetich JF, Hoareau-Coudert B, Mazier D, Silvie O. A rapid and robust selection procedure for generating drug-selectable marker-free recombinant malaria parasites. Sci Rep 2014; 4:4760. [PMID: 24755823 PMCID: PMC3996467 DOI: 10.1038/srep04760] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 04/04/2014] [Indexed: 12/22/2022] Open
Abstract
Experimental genetics have been widely used to explore the biology of the malaria parasites. The rodent parasites Plasmodium berghei and less frequently P. yoelii are commonly utilised, as their complete life cycle can be reproduced in the laboratory and because they are genetically tractable via homologous recombination. However, due to the limited number of drug-selectable markers, multiple modifications of the parasite genome are difficult to achieve and require large numbers of mice. Here we describe a novel strategy that combines positive-negative drug selection and flow cytometry-assisted sorting of fluorescent parasites for the rapid generation of drug-selectable marker-free P. berghei and P. yoelii mutant parasites expressing a GFP or a GFP-luciferase cassette, using minimal numbers of mice. We further illustrate how this new strategy facilitates phenotypic analysis of genetically modified parasites by fluorescence and bioluminescence imaging of P. berghei mutants arrested during liver stage development.
Collapse
Affiliation(s)
- Giulia Manzoni
- 1] Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France [2] INSERM, U1135, CIMI-Paris, 75013, Paris, France [3] CNRS, ERL 8255, CIMI-Paris, 75013, Paris, France [4]
| | - Sylvie Briquet
- 1] Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France [2] INSERM, U1135, CIMI-Paris, 75013, Paris, France [3] CNRS, ERL 8255, CIMI-Paris, 75013, Paris, France [4]
| | - Veronica Risco-Castillo
- 1] Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France [2] INSERM, U1135, CIMI-Paris, 75013, Paris, France [3] CNRS, ERL 8255, CIMI-Paris, 75013, Paris, France
| | - Charlotte Gaultier
- 1] Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France [2] INSERM, U1135, CIMI-Paris, 75013, Paris, France [3] CNRS, ERL 8255, CIMI-Paris, 75013, Paris, France [4]
| | - Selma Topçu
- 1] Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France [2] INSERM, U1135, CIMI-Paris, 75013, Paris, France [3] CNRS, ERL 8255, CIMI-Paris, 75013, Paris, France
| | - Maria Larisa Ivănescu
- 1] Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France [2] INSERM, U1135, CIMI-Paris, 75013, Paris, France [3] CNRS, ERL 8255, CIMI-Paris, 75013, Paris, France
| | - Jean-François Franetich
- 1] Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France [2] INSERM, U1135, CIMI-Paris, 75013, Paris, France [3] CNRS, ERL 8255, CIMI-Paris, 75013, Paris, France
| | - Bénédicte Hoareau-Coudert
- Sorbonne Universités, UPMC Univ Paris 06, Plateforme de Cytométrie en Flux CyPS, site Pitié-Salpêtrière, Paris, France
| | - Dominique Mazier
- 1] Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France [2] INSERM, U1135, CIMI-Paris, 75013, Paris, France [3] CNRS, ERL 8255, CIMI-Paris, 75013, Paris, France [4] Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Parasitologie-Mycologie, Paris, France
| | - Olivier Silvie
- 1] Sorbonne Universités, UPMC Univ Paris 06, CR7, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), 75013, Paris, France [2] INSERM, U1135, CIMI-Paris, 75013, Paris, France [3] CNRS, ERL 8255, CIMI-Paris, 75013, Paris, France
| |
Collapse
|
45
|
Chromosomal insertions in the Lactobacillus casei upp gene that are useful for vaccine expression. Appl Environ Microbiol 2014; 80:3321-6. [PMID: 24657853 DOI: 10.1128/aem.00175-14] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
To develop a stable and marker-free Lactobacillus strain useful for the expression of vaccines, we developed a temperature-sensitive suicide plasmid with expression cassettes containing an HCE promoter, a PgsA anchor, the alpha-toxin gene, and an rrnB T1T2 terminator (PPαT) that uses a 5-fluorouracil (5-FU) counterselectable marker for Lactobacillus casei. Three strains containing the correct PPαT expression cassettes were produced via the selective pressure of 5-FU screening. We confirmed that the upp gene was deleted and that the PPαT expression cassettes were inserted into the upp site of L. casei ATCC 393 by genomic PCR amplification and sequencing. 5-FU resistance in recombinant bacteria could be stably inherited for as long as 40 generations following insertion. However, bacteria containing the integrated DNA grew more slowly than wild-type L. casei. An indirect enzyme-linked immunosorbent assay (ELISA) analysis demonstrated that the alpha-toxin gene was expressed. Also, we visualized expression of the protein on the surface of L. casei cells using laser confocal microscopy. These results taken together demonstrate that these recombinant bacteria should provide a safe tool for effective vaccine production.
Collapse
|
46
|
Frénal K, Tay CL, Mueller C, Bushell ES, Jia Y, Graindorge A, Billker O, Rayner JC, Soldati-Favre D. Global analysis of apicomplexan protein S-acyl transferases reveals an enzyme essential for invasion. Traffic 2013; 14:895-911. [PMID: 23638681 PMCID: PMC3813974 DOI: 10.1111/tra.12081] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 04/29/2013] [Accepted: 05/02/2013] [Indexed: 11/27/2022]
Abstract
The advent of techniques to study palmitoylation on a whole proteome scale has revealed that it is an important reversible modification that plays a role in regulating multiple biological processes. Palmitoylation can control the affinity of a protein for lipid membranes, which allows it to impact protein trafficking, stability, folding, signalling and interactions. The publication of the palmitome of the schizont stage of Plasmodium falciparum implicated a role for palmitoylation in host cell invasion, protein export and organelle biogenesis. However, nothing is known so far about the repertoire of protein S-acyl transferases (PATs) that catalyse this modification in Apicomplexa. We undertook a comprehensive analysis of the repertoire of Asp-His-His-Cys cysteine-rich domain (DHHC-CRD) PAT family in Toxoplasma gondii and Plasmodium berghei by assessing their localization and essentiality. Unlike functional redundancies reported in other eukaryotes, some apicomplexan-specific DHHCs are essential for parasite growth, and several are targeted to organelles unique to this phylum. Of particular interest is DHHC7, which localizes to rhoptry organelles in all parasites tested, including the major human pathogen P. falciparum. TgDHHC7 interferes with the localization of the rhoptry palmitoylated protein TgARO and affects the apical positioning of the rhoptry organelles. This PAT has a major impact on T. gondii host cell invasion, but not on the parasite's ability to egress.
Collapse
Affiliation(s)
- Karine Frénal
- Department of Microbiology and Molecular Medicine, CMU, University of GenevaRue Michel-Servet 1, CH-1211, Geneva 4, Switzerland
| | - Chwen L Tay
- Malaria Programme, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, HinxtonCambridge, CB10 1SA, UK
| | - Christina Mueller
- Department of Microbiology and Molecular Medicine, CMU, University of GenevaRue Michel-Servet 1, CH-1211, Geneva 4, Switzerland
| | - Ellen S Bushell
- Malaria Programme, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, HinxtonCambridge, CB10 1SA, UK
| | - Yonggen Jia
- Department of Microbiology and Molecular Medicine, CMU, University of GenevaRue Michel-Servet 1, CH-1211, Geneva 4, Switzerland
| | - Arnault Graindorge
- Department of Microbiology and Molecular Medicine, CMU, University of GenevaRue Michel-Servet 1, CH-1211, Geneva 4, Switzerland
| | - Oliver Billker
- Malaria Programme, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, HinxtonCambridge, CB10 1SA, UK
| | - Julian C Rayner
- Malaria Programme, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, HinxtonCambridge, CB10 1SA, UK
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, CMU, University of GenevaRue Michel-Servet 1, CH-1211, Geneva 4, Switzerland
| |
Collapse
|
47
|
The exported protein PbCP1 localises to cleft-like structures in the rodent malaria parasite Plasmodium berghei. PLoS One 2013; 8:e61482. [PMID: 23658610 PMCID: PMC3637216 DOI: 10.1371/journal.pone.0061482] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 03/11/2013] [Indexed: 02/02/2023] Open
Abstract
Protein export into the host red blood cell is one of the key processes in the pathobiology of the malaria parasite Plasmodiumtrl falciparum, which extensively remodels the red blood cell to ensure its virulence and survival. In this study, we aimed to shed further light on the protein export mechanisms in the rodent malaria parasite P. berghei and provide further proof of the conserved nature of host cell remodeling in Plasmodium spp. Based on the presence of an export motif (R/KxLxE/Q/D) termed PEXEL (Plasmodium export element), we have generated transgenic P. berghei parasite lines expressing GFP chimera of putatively exported proteins and analysed one of the newly identified exported proteins in detail. This essential protein, termed PbCP1 (P. berghei Cleft-like Protein 1), harbours an atypical PEXEL motif (RxLxY) and is further characterised by two predicted transmembrane domains (2TMD) in the C-terminal end of the protein. We have functionally validated the unusual PEXEL motif in PbCP1 and analysed the role of the 2TMD region, which is required to recruit PbCP1 to discrete membranous structures in the red blood cell cytosol that have a convoluted, vesico-tubular morphology by electron microscopy. Importantly, this study reveals that rodent malaria species also induce modifications to their host red blood cell.
Collapse
|
48
|
Pasini EM, Braks JA, Fonager J, Klop O, Aime E, Spaccapelo R, Otto TD, Berriman M, Hiss JA, Thomas AW, Mann M, Janse CJ, Kocken CHM, Franke-Fayard B. Proteomic and genetic analyses demonstrate that Plasmodium berghei blood stages export a large and diverse repertoire of proteins. Mol Cell Proteomics 2012. [PMID: 23197789 PMCID: PMC3567864 DOI: 10.1074/mcp.m112.021238] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Malaria parasites actively remodel the infected red blood cell (irbc) by exporting proteins into the host cell cytoplasm. The human parasite Plasmodium falciparum exports particularly large numbers of proteins, including proteins that establish a vesicular network allowing the trafficking of proteins onto the surface of irbcs that are responsible for tissue sequestration. Like P. falciparum, the rodent parasite P. berghei ANKA sequesters via irbc interactions with the host receptor CD36. We have applied proteomic, genomic, and reverse-genetic approaches to identify P. berghei proteins potentially involved in the transport of proteins to the irbc surface. A comparative proteomics analysis of P. berghei non-sequestering and sequestering parasites was used to determine changes in the irbc membrane associated with sequestration. Subsequent tagging experiments identified 13 proteins (Plasmodium export element (PEXEL)-positive as well as PEXEL-negative) that are exported into the irbc cytoplasm and have distinct localization patterns: a dispersed and/or patchy distribution, a punctate vesicle-like pattern in the cytoplasm, or a distinct location at the irbc membrane. Members of the PEXEL-negative BIR and PEXEL-positive Pb-fam-3 show a dispersed localization in the irbc cytoplasm, but not at the irbc surface. Two of the identified exported proteins are transported to the irbc membrane and were named erythrocyte membrane associated proteins. EMAP1 is a member of the PEXEL-negative Pb-fam-1 family, and EMAP2 is a PEXEL-positive protein encoded by a single copy gene; neither protein plays a direct role in sequestration. Our observations clearly indicate that P. berghei traffics a diverse range of proteins to different cellular locations via mechanisms that are analogous to those employed by P. falciparum. This information can be exploited to generate transgenic humanized rodent P. berghei parasites expressing chimeric P. berghei/P. falciparum proteins on the surface of rodent irbc, thereby opening new avenues for in vivo screening adjunct therapies that block sequestration.
Collapse
Affiliation(s)
- Erica M Pasini
- Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Kooij TW, Rauch MM, Matuschewski K. Expansion of experimental genetics approaches for Plasmodium berghei with versatile transfection vectors. Mol Biochem Parasitol 2012; 185:19-26. [DOI: 10.1016/j.molbiopara.2012.06.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 05/26/2012] [Accepted: 06/05/2012] [Indexed: 11/28/2022]
|
50
|
Reininger L, Garcia M, Tomlins A, Müller S, Doerig C. The Plasmodium falciparum, Nima-related kinase Pfnek-4: a marker for asexual parasites committed to sexual differentiation. Malar J 2012; 11:250. [PMID: 22849771 PMCID: PMC3495404 DOI: 10.1186/1475-2875-11-250] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 07/13/2012] [Indexed: 12/15/2022] Open
Abstract
Background Malaria parasites undergo, in the vertebrate host, a developmental switch from asexual replication to sexual differentiation leading to the formation of gametocytes, the only form able to survive in the mosquito vector. Regulation of the onset of the sexual phase remains largely unknown and represents an important gap in the understanding of the parasite’s complex biology. Methods The expression and function of the Nima-related kinase Pfnek-4 during the early sexual development of the human malaria parasite Plasmodium falciparum were investigated, using three types of transgenic Plasmodium falciparum 3D7 lines: (i) episomally expressing a Pfnek-4-GFP fusion protein under the control of its cognate pfnek-4 promoter; (ii) episomally expressing negative or positive selectable markers, yeast cytosine deaminase-uridyl phosphoribosyl transferase, or human dihydrofolate reductase, under the control of the pfnek-4 promoter; and (iii) lacking a functional pfnek-4 gene. Parasite transfectants were analysed by fluorescence microscopy and flow cytometry. In vitro growth rate and gametocyte formation were determined by Giemsa-stained blood smears. Results The Pfnek-4-GFP protein was found to be expressed in stage II to V gametocytes and, unexpectedly, in a subset of asexual-stage parasites undergoing schizogony. Culture conditions stimulating gametocyte formation resulted in significant increase of this schizont subpopulation. Moreover, sorted asexual parasites expressing the Pfnek-4-GFP protein displayed elevated gametocyte formation when returned to in vitro culture in presence of fresh red blood cells, when compared to GFP- parasites from the same initial population. Negative selection of asexual parasites expressing pfnek-4 showed a marginal reduction in growth rate, whereas positive selection caused a marked reduction in parasitaemia, but was not sufficient to completely abolish proliferation. Pfnek-4- clones are not affected in their asexual growth and produced normal numbers of stage V gametocytes. Conclusions The results indicate that Pfnek-4 is not strictly gametocyte-specific, and is expressed in a small subset of asexual parasites displaying high rate conversion to sexual development. Pfnek-4 is not required for erythrocytic schizogony and gametocytogenesis. This is the first study to report the use of a molecular marker for the sorting of sexually-committed schizont stage P. falciparum parasites, which opens the way to molecular characterization of this pre-differentiated subpopulation.
Collapse
Affiliation(s)
- Luc Reininger
- CNRS USR3151, Station Biologique, Place Georges Teissier, 29680, Roscoff, France.
| | | | | | | | | |
Collapse
|