1
|
Atchou K, Caldelari R, Roques M, Schmuckli-Maurer J, Beyeler R, Heussler V. Expanding the fluorescent toolkit: Blue fluorescent protein-expressing Plasmodium berghei for enhanced multiplex microscopy. PLoS One 2025; 20:e0308055. [PMID: 40029851 PMCID: PMC11875362 DOI: 10.1371/journal.pone.0308055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 12/03/2024] [Indexed: 03/06/2025] Open
Abstract
Fluorescent proteins are widely used as markers to differentiate genetically modified cells from their wild-type counterparts. In malaria research, the prevalent fluorescent markers include red fluorescent proteins (RFPs) and their derivatives, such as mCherry, along with green fluorescent proteins (GFPs) and their derivatives. Recognizing the need for additional fluorescent markers to facilitate multiplexed imaging, this study introduced parasite lines expressing blue fluorescent protein (BFP). These lines enable simultaneous microscopy studies of proteins tagged with GFP, RFP, or detected by fluorophore-labeled antibodies, enhancing the analysis of complex biological interactions. Expression of BFP throughout the parasite's life cycle was driven by the robust Hsp70 promoter, ensuring stable, detectable protein levels suitable for fluorescent light analysis methods, including flow cytometry and fluorescent microscopy. We generated two Plasmodium berghei (P. berghei) lines expressing cytosolic BFP through double crossover homologous recombination targeting the silent 230p locus: eBFP2 (PbeBFP2) and mTagBFP2 (PbmTagBFP2). We compared these transgenic lines to established mCherry-expressing parasites PbmCherryHsp70 (PbmCherry) across their life cycles. The PbmTagBFP2 parasites exhibited fluorescence approximately 4.5 times brighter than the PbeBFP2 parasites in most life cycle stages. Both BFP-expressing lines developed normally through the entire parasite life cycle, offering a valuable expansion to the toolkit for studying Plasmodium biology at the host-pathogen interface.
Collapse
Affiliation(s)
- Kodzo Atchou
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Reto Caldelari
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Magali Roques
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | | | - Raphael Beyeler
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Volker Heussler
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| |
Collapse
|
2
|
Muchaamba G, Venugopal K, Gächter B, Vogler B, Hetzel U, Albini S, Marti M. Avian malaria in a feral-pet pigeon: a case report. Malar J 2024; 23:294. [PMID: 39358742 PMCID: PMC11446001 DOI: 10.1186/s12936-024-05116-5] [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: 06/06/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Avian malaria is caused by diverse parasite species of the genus Plasmodium, and it affects various bird species. The occurrence of this disease in some wild bird species is sparsely documented due to the scarce availability of samples. Hence the pathogenicity in some hosts is not completely known. In addition, feral birds may act as reservoirs bridging the transmission cycle from wild migratory birds to domestic and zoo-kept bird species. CASE PRESENTATION An owner of pigeons adopted a feral pigeon (Columba livia forma domestica) and housed it together with his other pet-pigeons. The bird died unexpectedly a few weeks after a surgical procedure and necropsy revealed a severely anaemic carcass, with pale organs and hydropericardium. Histopathologic analysis revealed inflammatory infiltrates in the lung and liver, and monocytes and Kupffer cells contained haemozoin pigment indicative of phagocytosis of Plasmodium-infected erythrocytes. A high erythrocytic infection rate of 18% was evident in tissues and blood vessels in various organs. Furthermore, the thyroid had masses classified as thyroid carcinomas. Immunohistochemistry with anti- Plasmodium falciparum HSP70 antibody revealed positive signals in erythrocytes and intravascular leucocytes. Further microscopy analysis using a Hemacolor-stained impression smear revealed a high parasitaemia with an asynchronous infection showing all erythrocytic stages. Molecular diagnosis by PCR identified Plasmodium relictum, lineage GRW11 as the aetiological agent. The bird presented died most likely due to an acute infection as evidenced by the high blood parasitaemia, leading to major erythrocyte destruction. Further analyses of feral pigeons (n = 22) did not reveal any additional cases of Plasmodium infections. CONCLUSION This study reports the first mortality associated with P. relictum lineage GRW11. The study supports previous studies, suggesting that Plasmodium infections are not frequent in pigeons. Host conditions like immunosuppression due to the tumour may have influenced the infection outcome in this fatal case. Use of anti-P. falciparum HSP70 antibody for detection of P. relictum antigens for immune assays in blood and tissue samples will be a useful tool for future studies.
Collapse
Affiliation(s)
- Gillian Muchaamba
- Institute of Parasitology, Vetsuisse and Medical Faculty, University of Zurich, Zurich, Switzerland
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Kannan Venugopal
- Institute of Parasitology, Vetsuisse and Medical Faculty, University of Zurich, Zurich, Switzerland
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| | - Bettina Gächter
- Section for Poultry and Rabbit Diseases (NRGK), Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Barbara Vogler
- Section for Poultry and Rabbit Diseases (NRGK), Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Udo Hetzel
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Sarah Albini
- Section for Poultry and Rabbit Diseases (NRGK), Institute for Food Safety and Hygiene, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Matthias Marti
- Institute of Parasitology, Vetsuisse and Medical Faculty, University of Zurich, Zurich, Switzerland.
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK.
| |
Collapse
|
3
|
A member of the tryptophan-rich protein family is required for efficient sequestration of Plasmodium berghei schizonts. PLoS Pathog 2022; 18:e1010846. [PMID: 36126089 PMCID: PMC9524624 DOI: 10.1371/journal.ppat.1010846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/30/2022] [Accepted: 08/31/2022] [Indexed: 11/20/2022] Open
Abstract
Protein export and host membrane remodeling are crucial for multiple Plasmodium species to establish a niche in infected hosts. To better understand the contribution of these processes to successful parasite infection in vivo, we sought to find and characterize protein components of the intraerythrocytic Plasmodium berghei-induced membrane structures (IBIS) that form in the cytoplasm of infected erythrocytes. We identified proteins that immunoprecipitate with IBIS1, a signature member of the IBIS in P. berghei-infected erythrocytes. In parallel, we also report our data describing proteins that co-precipitate with the PTEX (Plasmodium translocon of exported proteins) component EXP2. To validate our findings, we examined the location of three candidate IBIS1-interactors that are conserved across multiple Plasmodium species, and we found they localized to IBIS in infected red blood cells and two further colocalized with IBIS1 in the liver-stage parasitophorous vacuole membrane. Successful gene deletion revealed that these two tryptophan-rich domain-containing proteins, termed here IPIS2 and IPIS3 (for intraerythrocytic Plasmodium-induced membrane structures), are required for efficient blood-stage growth. Erythrocytes infected with IPIS2-deficient schizonts in particular fail to bind CD36 as efficiently as wild-type P. berghei-infected cells and therefore fail to effectively sequester out of the circulating blood. Our findings support the idea that intra-erythrocytic membrane compartments are required across species for alterations of the host erythrocyte that facilitate interactions of infected cells with host tissues.
Collapse
|
4
|
Hentzschel F, Gibbins MP, Attipa C, Beraldi D, Moxon CA, Otto TD, Marti M. Host cell maturation modulates parasite invasion and sexual differentiation in Plasmodium berghei. SCIENCE ADVANCES 2022; 8:eabm7348. [PMID: 35476438 PMCID: PMC9045723 DOI: 10.1126/sciadv.abm7348] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 03/11/2022] [Indexed: 05/04/2023]
Abstract
Malaria remains a global health problem causing more than 400,000 deaths annually. Plasmodium parasites, the causative agents of malaria, replicate asexually in red blood cells (RBCs) of their vertebrate host, while a subset differentiates into sexual stages (gametocytes) for mosquito transmission. Parasite replication and gametocyte maturation in the erythropoietic niches of the bone marrow and spleen contribute to pathogenesis and drive transmission, but the mechanisms underlying this organ enrichment remain unknown. Here, we performed a comprehensive analysis of rodent P. berghei infection by flow cytometry and single-cell RNA sequencing. We identified CD71 as a host receptor for reticulocyte invasion and found that parasites metabolically adapt to the host cell environment. Transcriptional analysis and functional assays further revealed a nutrient-dependent tropism for gametocyte formation in reticulocytes. Together, we provide a thorough characterization of host-parasite interactions in erythropoietic niches and define host cell maturation state as the key driver of parasite adaptation.
Collapse
Affiliation(s)
- Franziska Hentzschel
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| | - Matthew P. Gibbins
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| | - Charalampos Attipa
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
- Department of Pathology, Kamuzu University of Health Sciences, Blantyre, Malawi
- Malawi-Liverpool-Wellcome Clinical Research Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Dario Beraldi
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| | - Christopher A. Moxon
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- Malawi-Liverpool-Wellcome Clinical Research Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
- Department of Paediatrics and Child Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Thomas D. Otto
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| | - Matthias Marti
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA
| |
Collapse
|
5
|
Sahu T, Gehrke EJ, Flores-Garcia Y, Mlambo G, Romano JD, Coppens I. Chemoprophylaxis vaccination with a Plasmodium liver stage autophagy mutant affords enhanced and long-lasting protection. NPJ Vaccines 2021; 6:98. [PMID: 34376691 PMCID: PMC8355287 DOI: 10.1038/s41541-021-00360-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 07/06/2021] [Indexed: 11/09/2022] Open
Abstract
Genetically attenuated sporozoite vaccines can elicit long-lasting protection against malaria but pose risks of breakthrough infection. Chemoprophylaxis vaccination (CVac) has proven to be the most effective vaccine strategy against malaria. Here, we demonstrate that a liver stage-specific autophagy mutant of Plasmodium berghei (ATG8 overexpressor), when used as a live vaccine under a CVac regimen, provides superior long-lasting protection, in both inbred and outbred mice, as compared to WT-CVac. Uniquely, the protection elicited by this mutant is predominantly dependent on a CD8+ T-cell response through an IFN-γ-independent mechanism and is associated with a stable population of antigen-experienced CD8+ T cells. Jointly, our findings support the exploitation of liver-stage mutants as vaccines under a CVac protocol. This vaccination strategy is also a powerful model to study the mechanisms of protective immunity and discover new protective antigens.
Collapse
Affiliation(s)
- Tejram Sahu
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Ella J Gehrke
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Godfree Mlambo
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Julia D Romano
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
6
|
Rodriguez MC, Martínez-Barnetche J, Lecona-Valera AN, Téllez-Sosa J, Argotte-Ramos RS, Alvarado-Delgado A, Ovilla MT, Saldaña-Navor V, Rodriguez MH. Expression of Heat shock protein 70 (Hsp70-1) in Plasmodium berghei ookinetes and its participation in midgut mosquito infection. Mol Biochem Parasitol 2020; 240:111337. [PMID: 33147473 DOI: 10.1016/j.molbiopara.2020.111337] [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: 05/19/2020] [Revised: 10/22/2020] [Accepted: 10/28/2020] [Indexed: 10/23/2022]
Abstract
The heat shock protein family 70 (Hsp70) comprises chaperone proteins that play major multiple roles in Plasmodium asexual and sexual development. In this study, we analyzed the expression of Hsp70-1 in gametocytes, gametes, zygotes, and its participation in ookinete formation and their transition into oocysts. A monoclonal antibody against recombinant Hsp70-1 revealed its presence in zygotes and micronemes of ookinetes. Compared to wild type parasites, Hsp70-1 knockout ookinetes produced fewer oocysts in Plasmodium-susceptible Anopheles albimanus mosquitoes. This may indicate a defective transformation of ookinetes into oocysts in the absence of Hsp70-1. The presence of this protein in micronemes suggests its participation in mosquito infection, probably aiding to the adequate structural conformation of proteins in charge of motility, recognition and invasion of the insect midgut epithelium.
Collapse
Affiliation(s)
- Maria Carmen Rodriguez
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Av. Universidad 655, C. P. 62100, Cuernavaca, Morelos, Mexico
| | - Jesús Martínez-Barnetche
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Av. Universidad 655, C. P. 62100, Cuernavaca, Morelos, Mexico
| | - Alba N Lecona-Valera
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Av. Universidad 655, C. P. 62100, Cuernavaca, Morelos, Mexico
| | - Juan Téllez-Sosa
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Av. Universidad 655, C. P. 62100, Cuernavaca, Morelos, Mexico
| | - Rocio S Argotte-Ramos
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Av. Universidad 655, C. P. 62100, Cuernavaca, Morelos, Mexico
| | - Alejandro Alvarado-Delgado
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Av. Universidad 655, C. P. 62100, Cuernavaca, Morelos, Mexico
| | - Marbella T Ovilla
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Av. Universidad 655, C. P. 62100, Cuernavaca, Morelos, Mexico
| | - Vianey Saldaña-Navor
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Av. Universidad 655, C. P. 62100, Cuernavaca, Morelos, Mexico
| | - Mario H Rodriguez
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Av. Universidad 655, C. P. 62100, Cuernavaca, Morelos, Mexico.
| |
Collapse
|
7
|
Gnangnon B, Peucelle V, Pierrot C. Differential Fractionation of Erythrocytes Infected by Plasmodium berghei. Bio Protoc 2020; 10:e3647. [PMID: 33659316 DOI: 10.21769/bioprotoc.3647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 11/02/2022] Open
Abstract
The study of host/pathogen interactions at the cellular level during Plasmodium intra-erythrocytic cycle requires differential extraction techniques aiming to analyze the different compartments of the infected cell. Various protocols have been proposed in the literature to study specific compartments and/or membranes in the infected erythrocyte. The task remains delicate despite the use of enzymes or detergents theoretically capable of degrading specific membranes inside the infected cell. The remit of this protocol is to propose a method to isolate the erythrocyte cytosol and ghosts from the other compartments of the infected cell via a percoll gradient. Also, the lysis of the erythrocyte membrane is done using equinatoxin II, which has proven to be more effective at erythrocyte lysis regardless of the cell infection status, compared to the commonly used streptolysin. The parasitophorous vacuole (PV) content is collected after saponin lysis, before recovering membrane and parasite cytosol proteins by Triton X-100 lysis. The lysates thus obtained are analyzed by Western blot to assess the accuracy of the various extraction steps. This protocol allows the separation of the host compartment from the parasite compartments (PV and parasite), leading to potential studies of host proteins as well as parasite proteins exported to the host cell.
Collapse
Affiliation(s)
- Bénédicte Gnangnon
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Véronique Peucelle
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Christine Pierrot
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| |
Collapse
|
8
|
Kenthirapalan S, Tran PN, Kooij TWA, Ridgway MC, Rauch M, Brown SHJ, Mitchell TW, Matuschewski K, Maier AG. Distinct adaptations of a gametocyte ABC transporter to murine and human Plasmodium parasites and its incompatibility in cross-species complementation. Int J Parasitol 2020; 50:511-522. [PMID: 32445722 DOI: 10.1016/j.ijpara.2020.03.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 11/18/2022]
Abstract
Parasites of the genus Plasmodium infect a wide range of mammalian hosts including humans, primates, bats and arboreal rodents. A hallmark of Plasmodium spp. is the very narrow host range, indicative of matching parasite-host coevolution. Accordingly, their respective genomes harbour many unique genes and gene families that typically encode proteins involved in host cell recognition and remodelling. Whether and to what extent conserved proteins that are shared across Plasmodium spp. also exert distinct species-specific roles remains largely untested. Here, we present detailed functional profiling of the female gametocyte-specific ATP-binding cassette transporter gABCG2 in the murine parasite Plasmodium berghei and compare our findings with data from the orthologous gene in the human parasite Plasmodium falciparum. We show that P. berghei gABCG2 is female-specific and continues to be expressed in zygotes and ookinetes. In contrast to a distinct localization to Iipid-rich gametocyte-specific spots as observed in P. falciparum, the murine malaria parasite homolog is found at the parasite plasma membrane. Plasmodium berghei lacking gABCG2 displays fast asexual blood-stage replication and increased proportions of female gametocytes, consistent with the corresponding P. falciparum knock-out phenotype. Strikingly, cross-species replacement of gABCG2 in either the murine or the human parasite did not restore normal growth rates. The lack of successful complementation despite high conservation across Plasmodium spp. is an indicator of distinct adaptations and tight parasite-host coevolution. Hence, incompatibility of conserved genes in closely related Plasmodium spp. might be more common than previously anticipated.
Collapse
Affiliation(s)
| | - Phuong N Tran
- Research School of Biology, The Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Taco W A Kooij
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Melanie C Ridgway
- Research School of Biology, The Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Manuel Rauch
- Parasitology Unit, Max Planck Institute for Infection Biology, 10117 Berlin, Germany; Dept. of Molecular Parasitology, Institute of Biology, Humboldt University, 10115 Berlin, Germany
| | - Simon H J Brown
- School of Biological Sciences, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Todd W Mitchell
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia; School of Medicine, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Kai Matuschewski
- Parasitology Unit, Max Planck Institute for Infection Biology, 10117 Berlin, Germany; Dept. of Molecular Parasitology, Institute of Biology, Humboldt University, 10115 Berlin, Germany.
| | - Alexander G Maier
- Research School of Biology, The Australian National University, Canberra, Australian Capital Territory 2601, Australia.
| |
Collapse
|
9
|
Hentzschel F, Mitesser V, Fraschka SAK, Krzikalla D, Carrillo EH, Berkhout B, Bártfai R, Mueller AK, Grimm D. Gene knockdown in malaria parasites via non-canonical RNAi. Nucleic Acids Res 2020; 48:e2. [PMID: 31680162 PMCID: PMC7145648 DOI: 10.1093/nar/gkz927] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 10/01/2019] [Accepted: 10/07/2019] [Indexed: 12/13/2022] Open
Abstract
The lack of endogenous RNAi machinery in the malaria parasite Plasmodium hampers gene annotation and hence antimalarial drug and vaccine development. Here, we engineered rodent Plasmodium berghei to express a minimal, non-canonical RNAi machinery that solely requires Argonaute 2 (Ago2) and a modified short hairpin RNA, so-called AgoshRNA. Using this strategy, we achieved robust and specific gene knockdown throughout the entire parasite life cycle. We also successfully silenced the endogenous gene perforin-like protein 2, phenocopying a full gene knockout. Transcriptionally restricting Ago2 expression to the liver stage further enabled us to perform a stage-specific gene knockout. The RNAi-competent Plasmodium lines reported here will be a valuable resource for loss-of-function phenotyping of the many uncharacterized genes of Plasmodium in low or high throughput, without the need to engineer the target gene locus. Thereby, our new strategy and transgenic Plasmodium lines will ultimately benefit the discovery of urgently needed antimalarial drug and vaccine candidates. Generally, the ability to render RNAi-negative organisms RNAi-competent by mere introduction of two components, Ago2 and AgoshRNA, is a unique paradigm that should find broad applicability in other species.
Collapse
Affiliation(s)
- Franziska Hentzschel
- Heidelberg University Hospital, Center for Infectious Diseases / Parasitology, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany.,Heidelberg University Hospital, Center for Infectious Diseases / Virology, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany.,BioQuant Center, Heidelberg University, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| | - Vera Mitesser
- Heidelberg University Hospital, Center for Infectious Diseases / Parasitology, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany.,Heidelberg University Hospital, Center for Infectious Diseases / Virology, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany.,BioQuant Center, Heidelberg University, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| | | | - Daria Krzikalla
- Heidelberg University Hospital, Center for Infectious Diseases / Parasitology, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany.,Heidelberg University Hospital, Center for Infectious Diseases / Virology, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany.,BioQuant Center, Heidelberg University, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany
| | - Elena Herrera Carrillo
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology, Meibergdreef 15, K3-110, 1105 AZ Amsterdam, The Netherlands
| | - Ben Berkhout
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology, Meibergdreef 15, K3-110, 1105 AZ Amsterdam, The Netherlands
| | - Richárd Bártfai
- Radboud University, Dept. of Molecular Biology, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Ann-Kristin Mueller
- Heidelberg University Hospital, Center for Infectious Diseases / Parasitology, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany.,German Center for Infection Research (DZIF), partner site Heidelberg
| | - Dirk Grimm
- Heidelberg University Hospital, Center for Infectious Diseases / Virology, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany.,BioQuant Center, Heidelberg University, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany.,German Center for Infection Research (DZIF), partner site Heidelberg.,German Center for Cardiovascular Research (DZHK), partner site Heidelberg
| |
Collapse
|
10
|
Siden-Kiamos I, Goosmann C, Buscaglia CA, Brinkmann V, Matuschewski K, Montagna GN. Polarization of MTIP is a signature of gliding locomotion in Plasmodium ookinetes and sporozoites. Mol Biochem Parasitol 2019; 235:111247. [PMID: 31874192 DOI: 10.1016/j.molbiopara.2019.111247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 11/17/2022]
Abstract
Gliding motility and cell invasion are essential for the successful transmission of Plasmodium parasites. These processes rely on an acto-myosin motor located underneath the parasite plasma membrane. The Myosin A-tail interacting protein (MTIP) connects the class XIV myosin A (MyoA) to the gliding-associated proteins and is essential for assembly of the motor at the inner membrane complex. Here, we assessed the subcellular localization of MTIP in Plasmodium berghei motile stages from wild-type parasites and mutants that lack MyoA or the small heat shock protein 20 (HSP20). We demonstrate that MTIP is recruited to the apical end of motile ookinetes independently of the presence of MyoA. We also show that infective sporozoites displayed a polarized MTIP distribution during gliding, and that this distribution was abrogated in mutant parasites with an aberrant locomotion.
Collapse
Affiliation(s)
- Inga Siden-Kiamos
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Hellas, 700 13 Heraklion, Crete, Greece
| | | | - Carlos A Buscaglia
- Instituto de Investigaciones Biotecnológicas 'Dr Rodolfo Ugalde' (IIBio), UNSAM-CONICET, 1650 San Martín, Buenos Aires, Argentina
| | - Volker Brinkmann
- Max Planck Institute for Infection Biology, 10117 Berlin, Germany
| | - Kai Matuschewski
- Max Planck Institute for Infection Biology, 10117 Berlin, Germany; Department of Molecular Parasitology, Institute of Biology, Humboldt University, 10117 Berlin, Germany
| | - Georgina N Montagna
- Max Planck Institute for Infection Biology, 10117 Berlin, Germany; Instituto de Investigaciones Biotecnológicas 'Dr Rodolfo Ugalde' (IIBio), UNSAM-CONICET, 1650 San Martín, Buenos Aires, Argentina; Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, 049032, SP, Brazil..
| |
Collapse
|
11
|
Caldelari R, Dogga S, Schmid MW, Franke-Fayard B, Janse CJ, Soldati-Favre D, Heussler V. Transcriptome analysis of Plasmodium berghei during exo-erythrocytic development. Malar J 2019; 18:330. [PMID: 31551073 PMCID: PMC6760107 DOI: 10.1186/s12936-019-2968-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/17/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The complex life cycle of malaria parasites requires well-orchestrated stage specific gene expression. In the vertebrate host the parasites grow and multiply by schizogony in two different environments: within erythrocytes and within hepatocytes. Whereas erythrocytic parasites are well-studied in this respect, relatively little is known about the exo-erythrocytic stages. METHODS In an attempt to fill this gap, genome wide RNA-seq analyses of various exo-erythrocytic stages of Plasmodium berghei including sporozoites, samples from a time-course of liver stage development and detached cells were performed. These latter contain infectious merozoites and represent the final step in exo-erythrocytic development. RESULTS The analysis represents the complete transcriptome of the entire life cycle of P. berghei parasites with temporal detailed analysis of the liver stage allowing comparison of gene expression across the progression of the life cycle. These RNA-seq data from different developmental stages were used to cluster genes with similar expression profiles, in order to infer their functions. A comparison with published data from other parasite stages confirmed stage-specific gene expression and revealed numerous genes that are expressed differentially in blood and exo-erythrocytic stages. One of the most exo-erythrocytic stage-specific genes was PBANKA_1003900, which has previously been annotated as a "gametocyte specific protein". The promoter of this gene drove high GFP expression in exo-erythrocytic stages, confirming its expression profile seen by RNA-seq. CONCLUSIONS The comparative analysis of the genome wide mRNA expression profiles of erythrocytic and different exo-erythrocytic stages could be used to improve the understanding of gene regulation in Plasmodium parasites and can be used to model exo-erythrocytic stage metabolic networks toward the identification of differences in metabolic processes during schizogony in erythrocytes and hepatocytes.
Collapse
Affiliation(s)
- Reto Caldelari
- Institute of Cell Biology, University of Bern, Bern, Switzerland.
| | - Sunil Dogga
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva CMU, Geneva, Switzerland
| | | | - Blandine Franke-Fayard
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Chris J Janse
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva CMU, Geneva, Switzerland
| | - Volker Heussler
- Institute of Cell Biology, University of Bern, Bern, Switzerland.
| |
Collapse
|
12
|
Wells MB, Andrew DJ. Anopheles Salivary Gland Architecture Shapes Plasmodium Sporozoite Availability for Transmission. mBio 2019; 10:e01238-19. [PMID: 31387905 PMCID: PMC6686039 DOI: 10.1128/mbio.01238-19] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 07/11/2019] [Indexed: 12/18/2022] Open
Abstract
Plasmodium sporozoites (SPZs) must traverse the mosquito salivary glands (SGs) to reach a new vertebrate host and continue the malaria disease cycle. Although SGs can harbor thousands of sporozoites, only 10 to 100 are deposited into a host during probing. To determine how the SGs might function as a bottleneck in SPZ transmission, we have characterized Anopheles stephensi SGs infected with the rodent malaria parasite Plasmodium berghei using immunofluorescence confocal microscopy. Our analyses corroborate findings from previous electron microscopy studies and provide new insights into the invasion process. We identified sites of SPZ accumulation within SGs across a range of infection intensities. Although SPZs were most often seen in the distal lateral SG lobes, they were also observed in the medial and proximal lateral lobes. Most parasites were associated with either the basement membrane or secretory cavities. SPZs accumulated at physical barriers, including fused salivary ducts and extensions of the chitinous salivary duct wall into the distal lumen. SPZs were observed only rarely within salivary ducts. SPZs appeared to contact each other in many different quantities, not just in the previously described large bundles. Within parasite bundles, all of the SPZs were oriented in the same direction. We found that moderate levels of infection did not necessarily correlate with major SG disruptions or abundant SG cell death. Altogether, our findings suggest that SG architecture largely acts as a barrier to SPZ transmission.IMPORTANCE Malaria continues to have a devastating impact on human health. With growing resistance to insecticides and antimalarial drugs, as well as climate change predictions indicating expansion of vector territories, the impact of malaria is likely to increase. Additional insights regarding pathogen migration through vector mosquitoes are needed to develop novel methods to prevent transmission to new hosts. Pathogens, including the microbes that cause malaria, must invade the salivary glands (SGs) for transmission. Since SG traversal is required for parasite transmission, SGs are ideal targets for transmission-blocking strategies. The work presented here highlights the role that mosquito SG architecture plays in limiting parasite traversal, revealing how the SG transmission bottleneck is imposed. Further, our data provide unprecedented detail about SG-sporozoite interactions and gland-to-gland variation not provided in previous studies.
Collapse
Affiliation(s)
- Michael B Wells
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Deborah J Andrew
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
13
|
Alternative splicing is required for stage differentiation in malaria parasites. Genome Biol 2019; 20:151. [PMID: 31370870 PMCID: PMC6669979 DOI: 10.1186/s13059-019-1756-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 07/08/2019] [Indexed: 11/10/2022] Open
Abstract
Background In multicellular organisms, alternative splicing is central to tissue differentiation and identity. Unicellular protists lack multicellular tissue but differentiate into variable cell types during their life cycles. The role of alternative splicing in transitions between cell types and establishing cellular identity is currently unknown in any unicellular organism. Results To test whether alternative splicing in unicellular protists plays a role in cellular differentiation, we conduct RNA-seq to compare splicing in female and male sexual stages to asexual intraerythrocytic stages in the rodent malaria parasite Plasmodium berghei. We find extensive changes in alternative splicing between stages and a role for alternative splicing in sexual differentiation. Previously, general gametocyte differentiation was shown to be modulated by specific transcription factors. Here, we show that alternative splicing establishes a subsequent layer of regulation, controlling genes relating to consequent sex-specific differentiation of gametocytes. Conclusions We demonstrate that alternative splicing is reprogrammed during cellular differentiation of a unicellular protist. Disruption of an alternative splicing factor, PbSR-MG, perturbs sex-specific alternative splicing and decreases the ability of the parasites to differentiate into male gametes and oocysts, thereby reducing transmission between vertebrate and insect hosts. Our results reveal alternative splicing as an integral, stage-specific phenomenon in these protists and as a regulator of cellular differentiation that arose early in eukaryotic evolution. Electronic supplementary material The online version of this article (10.1186/s13059-019-1756-6) contains supplementary material, which is available to authorized users.
Collapse
|
14
|
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
|
15
|
Distinct Prominent Roles for Enzymes of Plasmodium berghei Heme Biosynthesis in Sporozoite and Liver Stage Maturation. Infect Immun 2016; 84:3252-3262. [PMID: 27600503 DOI: 10.1128/iai.00148-16] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 08/29/2016] [Indexed: 11/20/2022] Open
Abstract
Malarial parasites have evolved complex regulation of heme supply and disposal to adjust to heme-rich and -deprived host environments. In addition to its own pathway for heme biosynthesis, Plasmodium likely harbors mechanisms for heme scavenging from host erythrocytes. Elaborate compartmentalization of de novo heme synthesis into three subcellular locations, including the vestigial plastid organelle, indicates critical roles in life cycle progression. In this study, we systematically profile the essentiality of heme biosynthesis by targeted gene deletion of enzymes in early steps of this pathway. We show that disruption of endogenous heme biosynthesis leads to a first detectable defect in oocyst maturation and sporogony in the Anopheles vector, whereas blood stage propagation, colonization of mosquito midguts, or initiation of oocyst development occurs indistinguishably from that of wild-type parasites. Although sporozoites are produced by parasites lacking an intact pathway for heme biosynthesis, they are absent from mosquito salivary glands, indicative of a vital role for heme biosynthesis only in sporozoite maturation. Rescue of the first defect in sporogony permitted analysis of potential roles in liver stages. We show that liver stage parasites benefit from but do not strictly depend upon their own aminolevulinic acid synthase and that they can scavenge aminolevulinic acid from the host environment. Together, our experimental genetics analysis of Plasmodium enzymes for heme biosynthesis exemplifies remarkable shifts between the use of endogenous and host resources during life cycle progression.
Collapse
|
16
|
Sato Y, Hliscs M, Dunst J, Goosmann C, Brinkmann V, Montagna GN, Matuschewski K. Comparative Plasmodium gene overexpression reveals distinct perturbation of sporozoite transmission by profilin. Mol Biol Cell 2016; 27:2234-44. [PMID: 27226484 PMCID: PMC4945141 DOI: 10.1091/mbc.e15-10-0734] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 05/16/2016] [Indexed: 12/27/2022] Open
Abstract
The roles of vital genes, such as those of G-actin–binding proteins, in malaria parasites are underexplored. Overexpression of Plasmodium profilin perturbs actin dynamics only in sporozoites. Strict actin regulation is particularly important for malaria transmission. Mapping of phenotypes can be done by comparative Plasmodium gene overexpression. Plasmodium relies on actin-based motility to migrate from the site of infection and invade target cells. Using a substrate-dependent gliding locomotion, sporozoites are able to move at fast speed (1–3 μm/s). This motility relies on a minimal set of actin regulatory proteins and occurs in the absence of detectable filamentous actin (F-actin). Here we report an overexpression strategy to investigate whether perturbations of F-actin steady-state levels affect gliding locomotion and host invasion. We selected two vital Plasmodium berghei G-actin–binding proteins, C-CAP and profilin, in combination with three stage-specific promoters and mapped the phenotypes afforded by overexpression in all three extracellular motile stages. We show that in merozoites and ookinetes, additional expression does not impair life cycle progression. In marked contrast, overexpression of C-CAP and profilin in sporozoites impairs circular gliding motility and salivary gland invasion. The propensity for productive motility correlates with actin accumulation at the parasite tip, as revealed by combinations of an actin-stabilizing drug and transgenic parasites. Strong expression of profilin, but not C-CAP, resulted in complete life cycle arrest. Comparative overexpression is an alternative experimental genetic strategy to study essential genes and reveals effects of regulatory imbalances that are not uncovered from deletion-mutant phenotyping.
Collapse
Affiliation(s)
- Yuko Sato
- Parasitology Unit, Max Planck Institute for Infection Biology, 10117 Berlin, Germany Infectious Diseases Interdisciplinary Research Group, Singapore-Massachusetts Institute of Technology Alliance for Research and Technology, 138602 Singapore
| | - Marion Hliscs
- Parasitology Unit, Max Planck Institute for Infection Biology, 10117 Berlin, Germany School of BioSciences, University of Melbourne, Parkville, 3010 Victoria, Australia
| | - Josefine Dunst
- Parasitology Unit, Max Planck Institute for Infection Biology, 10117 Berlin, Germany Institute for Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Christian Goosmann
- Imaging Unit, Max Planck Institute for Infection Biology, 10117 Berlin, Germany
| | - Volker Brinkmann
- Imaging Unit, Max Planck Institute for Infection Biology, 10117 Berlin, Germany
| | - Georgina N Montagna
- Parasitology Unit, Max Planck Institute for Infection Biology, 10117 Berlin, Germany Departamento de Microbiologia, Immunologia e Parasitologia, Universidade Federal de São Paulo, 04039-032 São Paulo, Brazil
| | - Kai Matuschewski
- Parasitology Unit, Max Planck Institute for Infection Biology, 10117 Berlin, Germany Institute of Biology, Humboldt University, 10117 Berlin, Germany
| |
Collapse
|
17
|
Kalanon M, Bargieri D, Sturm A, Matthews K, Ghosh S, Goodman CD, Thiberge S, Mollard V, McFadden GI, Ménard R, Koning‐Ward TF. The
Plasmodium
translocon of exported proteins component EXP2 is critical for establishing a patent malaria infection in mice. Cell Microbiol 2015; 18:399-412. [DOI: 10.1111/cmi.12520] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 08/26/2015] [Accepted: 08/31/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Ming Kalanon
- Molecular and Medical Research Unit, School of MedicineDeakin University Waurn Ponds Geelong Victoria 3216 Australia
| | - Daniel Bargieri
- Unité de Biologie et Génétique du PaludismeInstitut Pasteur Paris France
- Department of Parasitology, Institute of Biomedical SciencesUniversity of São Paulo São Paulo SP Brazil
| | - Angelika Sturm
- School of BioSciencesThe University of Melbourne Parkville Victoria 3010 Australia
| | - Kathryn Matthews
- Molecular and Medical Research Unit, School of MedicineDeakin University Waurn Ponds Geelong Victoria 3216 Australia
| | - Sreejoyee Ghosh
- Molecular and Medical Research Unit, School of MedicineDeakin University Waurn Ponds Geelong Victoria 3216 Australia
| | | | - Sabine Thiberge
- Unité de Biologie et Génétique du PaludismeInstitut Pasteur Paris France
| | - Vanessa Mollard
- School of BioSciencesThe University of Melbourne Parkville Victoria 3010 Australia
| | - Geoffrey I. McFadden
- School of BioSciencesThe University of Melbourne Parkville Victoria 3010 Australia
| | - Robert Ménard
- Unité de Biologie et Génétique du PaludismeInstitut Pasteur Paris France
| | - Tania F. Koning‐Ward
- Molecular and Medical Research Unit, School of MedicineDeakin University Waurn Ponds Geelong Victoria 3216 Australia
| |
Collapse
|
18
|
The subcellular location of ovalbumin in Plasmodium berghei blood stages influences the magnitude of T-cell responses. Infect Immun 2014; 82:4654-65. [PMID: 25156724 DOI: 10.1128/iai.01940-14] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Model antigens are frequently introduced into pathogens to study determinants that influence T-cell responses to infections. To address whether an antigen's subcellular location influences the nature and magnitude of antigen-specific T-cell responses, we generated Plasmodium berghei parasites expressing the model antigen ovalbumin (OVA) either in the parasite cytoplasm or on the parasitophorous vacuole membrane (PVM). For cytosolic expression, OVA alone or conjugated to mCherry was expressed from a strong constitutive promoter (OVAhsp70 or OVA::mCherryhsp70); for PVM expression, OVA was fused to HEP17/EXP1 (OVA::Hep17hep17). Unexpectedly, OVA expression in OVAhsp70 parasites was very low, but when OVA was fused to mCherry (OVA::mCherryhsp70), it was highly expressed. OVA expression in OVA::Hep17hep17 parasites was strong but significantly less than that in OVA::mCherryhsp70 parasites. These transgenic parasites were used to examine the effects of antigen subcellular location and expression level on the development of T-cell responses during blood-stage infections. While all OVA-expressing parasites induced activation and proliferation of OVA-specific CD8(+) T cells (OT-I) and CD4(+) T cells (OT-II), the level of activation varied: OVA::Hep17hep17 parasites induced significantly stronger splenic and intracerebral OT-I and OT-II responses than those of OVA::mCherryhsp70 parasites, but OVA::mCherryhsp70 parasites promoted stronger OT-I and OT-II responses than those of OVAhsp70 parasites. Despite lower OVA expression levels, OVA::Hep17hep17 parasites induced stronger T-cell responses than those of OVA::mCherryhsp70 parasites. These results indicate that unconjugated cytosolic OVA is not stably expressed in Plasmodium parasites and, importantly, that its cellular location and expression level influence both the induction and magnitude of parasite-specific T-cell responses. These parasites represent useful tools for studying the development and function of antigen-specific T-cell responses during malaria infection.
Collapse
|
19
|
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: 42] [Impact Index Per Article: 3.8] [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
|