1
|
Guizetti J. Imaging malaria parasites across scales and time. J Microsc 2025. [PMID: 39749880 DOI: 10.1111/jmi.13384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
The idea that disease is caused at the cellular level is so fundamental to us that we might forget the critical role microscopy played in generating and developing this insight. Visually identifying diseased or infected cells lays the foundation for any effort to curb human pathology. Since the discovery of the Plasmodium-infected red blood cells, which cause malaria, microscopy has undergone an impressive development now literally resolving individual molecules. This review explores the expansive field of light microscopy, focusing on its application to malaria research. Imaging technologies have transformed our understanding of biological systems, yet navigating the complex and ever-growing landscape of techniques can be daunting. This review offers a guide for researchers, especially those working on malaria, by providing historical context as well as practical advice on selecting the right imaging approach. The review advocates an integrated methodology that prioritises the research question while considering key factors like sample preparation, fluorophore choice, imaging modality, and data analysis. In addition to presenting seminal studies and innovative applications of microscopy, the review highlights a broad range of topics, from traditional techniques like white light microscopy to advanced methods such as superresolution microscopy and time-lapse imaging. It addresses the emerging challenges of microscopy, including phototoxicity and trade-offs in resolution and speed, and offers insights into future technologies that might impact malaria research. This review offers a mix of historical perspective, technological progress, and practical guidance that appeal to novice and advanced microscopists alike. It aims to inspire malaria researchers to explore imaging techniques that could enrich their studies, thus advancing the field through enhanced visual exploration of the parasite across scales and time.
Collapse
Affiliation(s)
- Julien Guizetti
- Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
2
|
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
|
3
|
Novobilský A, Höglund J. Small animal in vivo imaging of parasitic infections: A systematic review. Exp Parasitol 2020; 214:107905. [PMID: 32387050 DOI: 10.1016/j.exppara.2020.107905] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/27/2020] [Accepted: 05/01/2020] [Indexed: 10/24/2022]
Abstract
Non-invasive small animal in vivo imaging is an essential tool in a broad variety of biomedical sciences and enables continuous monitoring of disease progression in order to develop and improve diagnostic, therapeutic and preventive measures. Imaging parasites non-invasively in live animals allows efficient parasite distribution evaluation in the host organism and objective evaluation of parasitic diseases' burden and progression in individual animals. The aim of this systematic review was to summarize recent trends in small animal in vivo imaging and compare and discuss imaging of single-cell and multicellular eukaryotic parasites. A literature survey was performed using Web of Science and PubMed databases in research articles published between 1990 and 2018. The inclusion criteria were using any imaging method to visualize a range of protozoan and helminth parasites in laboratory animals in vivo. A total of 92 studies met our inclusion criteria. Protozoans and helminths were imaged in 88% and 12% of 92 studies, respectively. The most common parasite genus studied was the protozoan Plasmodium followed by Trypanosoma and Leishmania. The most frequent imaging method was bioluminescence. Among the helminths, Schistosoma and Echinococcus were the most studied organisms. In vivo imaging is applicable in both protozoans and helminths. In helminths, however, the use of in vivo imaging methods is limited to some extent. Imaging parasites in small animal models is a powerful tool in preclinical research aiming to develop novel therapeutic and preventive strategies for parasitic diseases of interest both in human and veterinary medicine.
Collapse
Affiliation(s)
- Adam Novobilský
- Veterinary Research Institute, Department of Pharmacology and Immunotherapy, Hudcova 70, 62100, Brno, Czech Republic.
| | - Johan Höglund
- Swedish University of Agricultural Sciences, Department of Biological Sciences and Veterinary Public Health, Section for Parasitology, Box 7036, 750 07, Uppsala, Sweden
| |
Collapse
|
4
|
Othman AS, Marin-Mogollon C, Salman AM, Franke-Fayard BM, Janse CJ, Khan SM. The use of transgenic parasites in malaria vaccine research. Expert Rev Vaccines 2017; 16:1-13. [DOI: 10.1080/14760584.2017.1333426] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Ahmad Syibli Othman
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
- Faculty of Health Sciences, Universiti Sultan Zainal Abidin, Terengganu, Malaysia
| | - Catherin Marin-Mogollon
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | | | - Blandine M. Franke-Fayard
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Chris J. Janse
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Shahid M. Khan
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| |
Collapse
|
5
|
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
|
6
|
Ferreira BL, Orikaza CM, Cordero EM, Mortara RA. Trypanosoma cruzi: single cell live imaging inside infected tissues. Cell Microbiol 2016; 18:779-83. [PMID: 26639617 PMCID: PMC5064609 DOI: 10.1111/cmi.12553] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 11/03/2015] [Accepted: 12/03/2015] [Indexed: 01/30/2023]
Abstract
Although imaging the live Trypanosoma cruzi parasite is a routine technique in most laboratories, identification of the parasite in infected tissues and organs has been hindered by their intrinsic opaque nature. We describe a simple method for in vivo observation of live single-cell Trypanosoma cruzi parasites inside mammalian host tissues. BALB/c or C57BL/6 mice infected with DsRed-CL or GFP-G trypomastigotes had their organs removed and sectioned with surgical blades. Ex vivo organ sections were observed under confocal microscopy. For the first time, this procedure enabled imaging of individual amastigotes, intermediate forms and motile trypomastigotes within infected tissues of mammalian hosts.
Collapse
Affiliation(s)
- Bianca Lima Ferreira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM-UNIFESP), Rua Botucatu 862 6th floor, São Paulo, São Paulo, Brazil
| | - Cristina Mary Orikaza
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM-UNIFESP), Rua Botucatu 862 6th floor, São Paulo, São Paulo, Brazil
| | - Esteban Mauricio Cordero
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM-UNIFESP), Rua Botucatu 862 6th floor, São Paulo, São Paulo, Brazil
| | - Renato Arruda Mortara
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM-UNIFESP), Rua Botucatu 862 6th floor, São Paulo, São Paulo, Brazil
| |
Collapse
|
7
|
Douglas RG, Amino R, Sinnis P, Frischknecht F. Active migration and passive transport of malaria parasites. Trends Parasitol 2015; 31:357-62. [PMID: 26001482 DOI: 10.1016/j.pt.2015.04.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 04/22/2015] [Accepted: 04/23/2015] [Indexed: 11/16/2022]
Abstract
Malaria parasites undergo a complex life cycle between their hosts and vectors. During this cycle the parasites invade different types of cells, migrate across barriers, and transfer from one host to another. Recent literature hints at a misunderstanding of the difference between active, parasite-driven migration and passive, circulation-driven movement of the parasite or parasite-infected cells in the various bodily fluids of mosquito and mammalian hosts. Because both active migration and passive transport could be targeted in different ways to interfere with the parasite, a distinction between the two ways the parasite uses to get from one location to another is essential. We discuss the two types of motion needed for parasite dissemination and elaborate on how they could be targeted by future vaccines or drugs.
Collapse
Affiliation(s)
- Ross G Douglas
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Rogerio Amino
- Unité de Biologie et Génétique du Paludisme, Département Parasites et Insectes Vecteurs, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Photini Sinnis
- Johns Hopkins Malaria Research Institute, Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Freddy Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany.
| |
Collapse
|
8
|
Frevert U, Nacer A. Immunobiology of Plasmodium in liver and brain. Parasite Immunol 2014; 35:267-82. [PMID: 23631610 DOI: 10.1111/pim.12039] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Accepted: 04/17/2013] [Indexed: 12/11/2022]
Abstract
Malaria remains one of the most serious health problems globally, but our understanding of the biology of the parasite and the pathogenesis of severe disease is still limited. Multiple cellular effector mechanisms that mediate parasite elimination from the liver have been described, but how effector cells use classical granule-mediated cytotoxicity to attack infected hepatocytes and how cytokines and chemokines spread via the unique fluid pathways of the liver to reach the parasites over considerable distances remains unknown. Similarly, a wealth of information on cerebral malaria (CM), one of the most severe manifestations of the disease, was gained from post-mortem analyses of human brain and murine disease models, but the cellular processes that ultimately cause disease are not fully understood. Here, we discuss how imaging of the local dynamics of parasite infection and host response as well as consideration of anatomical and physiological features of liver and brain can provide a better understanding of the initial asymptomatic hepatic phase of the infection and the cascade of events leading to CM. Given the increasing drug resistance of both parasite and vector and the unavailability of a protective vaccine, the urgency to reduce the tremendous morbidity and mortality associated with severe malaria is obvious.
Collapse
Affiliation(s)
- U Frevert
- Division of Medical Parasitology, Department of Microbiology, New York University School of Medicine, New York, NY 10010, USA.
| | | |
Collapse
|
9
|
Ferrer M, Martin-Jaular L, De Niz M, Khan SM, Janse CJ, Calvo M, Heussler V, del Portillo HA. Imaging of the spleen in malaria. Parasitol Int 2013; 63:195-205. [PMID: 23999413 DOI: 10.1016/j.parint.2013.08.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Revised: 08/13/2013] [Accepted: 08/26/2013] [Indexed: 11/28/2022]
Abstract
Splenomegaly, albeit variably, is a hallmark of malaria; yet, the role of the spleen in Plasmodium infections remains vastly unknown. The implementation of imaging to study the spleen is rapidly advancing our knowledge of this so-called "blackbox" of the abdominal cavity. Not only has ex vivo imaging revealed the complex functional compartmentalization of the organ and immune effector cells, but it has also allowed the observation of major structural remodeling during infections. In vivo imaging, on the other hand, has allowed quantitative measurements of the dynamic passage of the parasite at spatial and temporal resolution. Here, we review imaging techniques used for studying the malarious spleen, from optical microscopy to in vivo imaging, and discuss the bright perspectives of evolving technologies in our present understanding of the role of this organ in infections caused by Plasmodium.
Collapse
Affiliation(s)
- Mireia Ferrer
- Barcelona Centre for International Health Research (CRESIB, Hospital Clínic-Universitat de Barcelona) ISGlobal, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Kudryashev M, Münter S, Lemgruber L, Montagna G, Stahlberg H, Matuschewski K, Meissner M, Cyrklaff M, Frischknecht F. Structural basis for chirality and directional motility of Plasmodium sporozoites. Cell Microbiol 2012; 14:1757-68. [PMID: 22776715 DOI: 10.1111/j.1462-5822.2012.01836.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 06/28/2012] [Accepted: 07/03/2012] [Indexed: 11/28/2022]
Abstract
Plasmodium sporozoites can move at high speed for several tens of minutes, which is essential for the initial stage of a malaria infection. The crescent-shaped sporozoites move on 2D substrates preferably in the same direction on circular paths giving raise to helical paths in 3D matrices. Here we determined the structural basis that underlies this type of movement. Immature, non-motile sporozoites were found to lack the subpellicular network required for obtaining the crescent parasite shape. In vitro, parasites moving in the favoured direction move faster and more persistent than the few parasites that move in the opposite direction. Photobleaching experiments showed that sporozoites flip their ventral side up when switching the direction of migration. Cryo-electron tomography revealed a polarized arrangement of microtubules and polar rings towards the substrate in Plasmodium sporozoites, but not in the related parasite Toxoplasma gondii. As a consequence, secretory vesicles, which release proteins involved in adhesion, migration and invasion at the front end of the parasite, are delivered towards the substrate. The resulting chiral structure of the parasite appears to determine the unique directionality of movement and could explain how the sporozoite achieves rapid and sustained directional motility in the absence of external stimuli.
Collapse
Affiliation(s)
- Mikhail Kudryashev
- Parasitology, Department of Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Asada M, Goto Y, Yahata K, Yokoyama N, Kawai S, Inoue N, Kaneko O, Kawazu SI. Gliding motility of Babesia bovis merozoites visualized by time-lapse video microscopy. PLoS One 2012; 7:e35227. [PMID: 22506073 PMCID: PMC3323635 DOI: 10.1371/journal.pone.0035227] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 03/12/2012] [Indexed: 11/22/2022] Open
Abstract
Background Babesia bovis is an apicomplexan intraerythrocytic protozoan parasite that induces babesiosis in cattle after transmission by ticks. During specific stages of the apicomplexan parasite lifecycle, such as the sporozoites of Plasmodium falciparum and tachyzoites of Toxoplasma gondii, host cells are targeted for invasion using a unique, active process termed “gliding motility”. However, it is not thoroughly understood how the merozoites of B. bovis target and invade host red blood cells (RBCs), and gliding motility has so far not been observed in the parasite. Methodology/Principal Findings Gliding motility of B. bovis merozoites was revealed by time-lapse video microscopy. The recorded images revealed that the process included egress of the merozoites from the infected RBC, gliding motility, and subsequent invasion into new RBCs. The gliding motility of B. bovis merozoites was similar to the helical gliding of Toxoplasma tachyzoites. The trails left by the merozoites were detected by indirect immunofluorescence assay using antiserum against B. bovis merozoite surface antigen 1. Inhibition of gliding motility by actin filament polymerization or depolymerization indicated that the gliding motility was driven by actomyosin dependent process. In addition, we revealed the timing of breakdown of the parasitophorous vacuole. Time-lapse image analysis of membrane-stained bovine RBCs showed formation and breakdown of the parasitophorous vacuole within ten minutes of invasion. Conclusions/Significance This is the first report of the gliding motility of B. bovis. Since merozoites of Plasmodium parasites do not glide on a substrate, the gliding motility of B. bovis merozoites is a notable finding.
Collapse
Affiliation(s)
- Masahito Asada
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada, Obihiro, Japan
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Rénia L, Howland SW, Claser C, Charlotte Gruner A, Suwanarusk R, Hui Teo T, Russell B, Ng LFP. Cerebral malaria: mysteries at the blood-brain barrier. Virulence 2012; 3:193-201. [PMID: 22460644 PMCID: PMC3396698 DOI: 10.4161/viru.19013] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cerebral malaria is the most severe pathology caused by the malaria parasite, Plasmodium falciparum. The pathogenic mechanisms leading to cerebral malaria are still poorly defined as studies have been hampered by limited accessibility to human tissues. Nevertheless, histopathology of post-mortem human tissues and mouse models of cerebral malaria have indicated involvement of the blood-brain barrier in cerebral malaria. In contrast to viruses and bacteria, malaria parasites do not infiltrate and infect the brain parenchyma. Instead, rupture of the blood-brain barrier occurs and may lead to hemorrhages resulting in neurological alterations. Here, we review the most recent findings from human studies and mouse models on the interactions of malaria parasites and the blood-brain barrier, shedding light on the pathogenesis of cerebral malaria, which may provide directions for possible interventions.
Collapse
Affiliation(s)
- Laurent Rénia
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A STAR), Biopolis, Singapore.
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Ferrer M, Martin-Jaular L, Calvo M, del Portillo HA. Intravital microscopy of the spleen: quantitative analysis of parasite mobility and blood flow. J Vis Exp 2012:3609. [PMID: 22298018 DOI: 10.3791/3609] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The advent of intravital microscopy in experimental rodent malaria models has allowed major advances to the knowledge of parasite-host interactions. Thus, in vivo imaging of malaria parasites during pre-erythrocytic stages have revealed the active entrance of parasites into skin lymph nodes, the complete development of the parasite in the skin, and the formation of a hepatocyte-derived merosome to assure migration and release of merozoites into the blood stream. Moreover, the development of individual parasites in erythrocytes has been recently documented using 4D imaging and challenged our current view on protein export in malaria. Thus, intravital imaging has radically changed our view on key events in Plasmodium development. Unfortunately, studies of the dynamic passage of malaria parasites through the spleen, a major lymphoid organ exquisitely adapted to clear infected red blood cells are lacking due to technical constraints. Using the murine model of malaria Plasmodium yoelii in Balb/c mice, we have implemented intravital imaging of the spleen and reported a differential remodeling of it and adherence of parasitized red blood cells (pRBCs) to barrier cells of fibroblastic origin in the red pulp during infection with the non-lethal parasite line P.yoelii 17X as opposed to infections with the P.yoelii 17XL lethal parasite line. To reach these conclusions, a specific methodology using ImageJ free software was developed to enable characterization of the fast three-dimensional movement of single-pRBCs. Results obtained with this protocol allow determining velocity, directionality and residence time of parasites in the spleen, all parameters addressing adherence in vivo. In addition, we report the methodology for blood flow quantification using intravital microscopy and the use of different colouring agents to gain insight into the complex microcirculatory structure of the spleen. ETHICS STATEMENT: All the animal studies were performed at the animal facilities of University of Barcelona in accordance with guidelines and protocols approved by the Ethics Committee for Animal Experimentation of the University of Barcelona CEEA-UB (Protocol No DMAH: 5429). Female Balb/c mice of 6-8 weeks of age were obtained from Charles River Laboratories.
Collapse
Affiliation(s)
- Mireia Ferrer
- Department of poverty related diseases, Barcelona Centre for International Health Research
| | | | | | | |
Collapse
|
14
|
Abstract
Unicellular parasites are of high medical relevance as they cause such devastating diseases as malaria or sleeping sickness. Besides the search for improved treatments, research on these parasites is valuable as they constitute interesting model cells to study basic processes of life. They can also serve as valuable reality checks for our presumed understanding of biological processes that emerge from the study of human or yeast cells, as our common ancestor with many parasites is much older than the one with yeast. But working with parasites can be tricky and time-consuming, if not outright impossible. Here, we focus on examples from imaging studies investigating the transmission of the malaria parasite. Achieving an understanding of the processes important for malaria transmission necessitates different imaging approaches and new molecular and material technologies. The discussed techniques will include in vivo imaging of pathogens in living animals, screening methodologies, and new materials as surrogate 3D environments.
Collapse
Affiliation(s)
- Mirko Singer
- Department of Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | | |
Collapse
|
15
|
Lin JW, Annoura T, Sajid M, Chevalley-Maurel S, Ramesar J, Klop O, Franke-Fayard BMD, Janse CJ, Khan SM. A novel 'gene insertion/marker out' (GIMO) method for transgene expression and gene complementation in rodent malaria parasites. PLoS One 2011; 6:e29289. [PMID: 22216235 PMCID: PMC3246482 DOI: 10.1371/journal.pone.0029289] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 11/24/2011] [Indexed: 01/25/2023] Open
Abstract
Research on the biology of malaria parasites has greatly benefited from the application of reverse genetic technologies, in particular through the analysis of gene deletion mutants and studies on transgenic parasites that express heterologous or mutated proteins. However, transfection in Plasmodium is limited by the paucity of drug-selectable markers that hampers subsequent genetic modification of the same mutant. We report the development of a novel 'gene insertion/marker out' (GIMO) method for two rodent malaria parasites, which uses negative selection to rapidly generate transgenic mutants ready for subsequent modifications. We have created reference mother lines for both P. berghei ANKA and P. yoelii 17XNL that serve as recipient parasites for GIMO-transfection. Compared to existing protocols GIMO-transfection greatly simplifies and speeds up the generation of mutants expressing heterologous proteins, free of drug-resistance genes, and requires far fewer laboratory animals. In addition we demonstrate that GIMO-transfection is also a simple and fast method for genetic complementation of mutants with a gene deletion or mutation. The implementation of GIMO-transfection procedures should greatly enhance Plasmodium reverse-genetic research.
Collapse
Affiliation(s)
- Jing-wen Lin
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Takeshi Annoura
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Mohammed Sajid
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Séverine Chevalley-Maurel
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Jai Ramesar
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Onny Klop
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Blandine M. D. Franke-Fayard
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Chris J. Janse
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Shahid M. Khan
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Centre, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
16
|
A genotype and phenotype database of genetically modified malaria-parasites. Trends Parasitol 2010; 27:31-9. [PMID: 20663715 DOI: 10.1016/j.pt.2010.06.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2010] [Revised: 06/15/2010] [Accepted: 06/16/2010] [Indexed: 11/20/2022]
Abstract
The RMgm database, www.pberghei.eu, is a web-based, manually curated, repository containing information on genetically modified rodent-malaria parasites. It provides easy and rapid access to information on the genotype and phenotype of mutant and reporter parasites. The database also contains information on unpublished mutants without a clear phenotype and negative trials to disrupt genes. Information can be searched using pre-defined key features, such as phenotype, life-cycle stage, gene model, gene-tags and mutations. The information relating to the mutants is reciprocally linked to PlasmoDB and GeneDB. Access to mutant-parasite information, and gene function/ontology inferred from mutant phenotypes provides a timely resource aimed at enhancing research into Plasmodium gene function and (systems) biology.
Collapse
|
17
|
Hegge S, Munter S, Steinbüchel M, Heiss K, Engel U, Matuschewski K, Frischknecht F. Multistep adhesion of
Plasmodium
sporozoites. FASEB J 2010; 24:2222-34. [DOI: 10.1096/fj.09-148700] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Stephan Hegge
- Department of ParasitologyHygiene InstituteUniversity of Heidelberg Medical SchoolHeidelbergGermany
| | - Sylvia Munter
- Department of ParasitologyHygiene InstituteUniversity of Heidelberg Medical SchoolHeidelbergGermany
| | - Marion Steinbüchel
- Department of ParasitologyHygiene InstituteUniversity of Heidelberg Medical SchoolHeidelbergGermany
| | - Kirsten Heiss
- Department of ParasitologyHygiene InstituteUniversity of Heidelberg Medical SchoolHeidelbergGermany
| | | | - Kai Matuschewski
- Department of ParasitologyHygiene InstituteUniversity of Heidelberg Medical SchoolHeidelbergGermany
| | - Friedrich Frischknecht
- Department of ParasitologyHygiene InstituteUniversity of Heidelberg Medical SchoolHeidelbergGermany
| |
Collapse
|
18
|
Ploemen IHJ, Prudêncio M, Douradinha BG, Ramesar J, Fonager J, van Gemert GJ, Luty AJF, Hermsen CC, Sauerwein RW, Baptista FG, Mota MM, Waters AP, Que I, Lowik CWGM, Khan SM, Janse CJ, Franke-Fayard BMD. Visualisation and quantitative analysis of the rodent malaria liver stage by real time imaging. PLoS One 2009; 4:e7881. [PMID: 19924309 PMCID: PMC2775639 DOI: 10.1371/journal.pone.0007881] [Citation(s) in RCA: 199] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Accepted: 10/26/2009] [Indexed: 11/17/2022] Open
Abstract
The quantitative analysis of Plasmodium development in the liver in laboratory animals in cultured cells is hampered by low parasite infection rates and the complicated methods required to monitor intracellular development. As a consequence, this important phase of the parasite's life cycle has been poorly studied compared to blood stages, for example in screening anti-malarial drugs. Here we report the use of a transgenic P. berghei parasite, PbGFP-Luccon, expressing the bioluminescent reporter protein luciferase to visualize and quantify parasite development in liver cells both in culture and in live mice using real-time luminescence imaging. The reporter-parasite based quantification in cultured hepatocytes by real-time imaging or using a microplate reader correlates very well with established quantitative RT-PCR methods. For the first time the liver stage of Plasmodium is visualized in whole bodies of live mice and we were able to discriminate as few as 1–5 infected hepatocytes per liver in mice using 2D-imaging and to identify individual infected hepatocytes by 3D-imaging. The analysis of liver infections by whole body imaging shows a good correlation with quantitative RT-PCR analysis of extracted livers. The luminescence-based analysis of the effects of various drugs on in vitro hepatocyte infection shows that this method can effectively be used for in vitro screening of compounds targeting Plasmodium liver stages. Furthermore, by analysing the effect of primaquine and tafenoquine in vivo we demonstrate the applicability of real time imaging to assess parasite drug sensitivity in the liver. The simplicity and speed of quantitative analysis of liver-stage development by real-time imaging compared to the PCR methodologies, as well as the possibility to analyse liver development in live mice without surgery, opens up new possibilities for research on Plasmodium liver infections and for validating the effect of drugs and vaccines on the liver stage of Plasmodium.
Collapse
Affiliation(s)
- Ivo H J Ploemen
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre (RUNMC), Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Neither mosquito saliva nor immunity to saliva has a detectable effect on the infectivity of Plasmodium sporozoites injected into mice. Infect Immun 2009; 78:545-51. [PMID: 19884338 DOI: 10.1128/iai.00807-09] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Malaria infection is initiated when a female Anopheles mosquito probing for blood injects saliva, together with sporozoites, into the skin of its mammalian host. Prior studies had suggested that saliva may enhance sporozoite infectivity. Using rodent malaria models (Plasmodium berghei and P. yoelii), we were unable to show that saliva had any detectable effect on sporozoite infectivity. This is encouraging for plans to immunize humans with washed, attenuated P. falciparum sporozoites because many individuals develop cutaneous, hypersensitivity reactions to mosquito saliva after repeated exposure. If washed sporozoites have no appreciable loss of infectivity, they likely do not have decreased immunogenicity; thus, vaccinees are unlikely to develop cutaneous reactions against mosquito saliva during attempted immunization with such sporozoites. Earlier studies also suggested that repeated prior exposure to mosquito saliva reduces infectivity of sporozoites injected by mosquitoes into sensitized hosts. However, our own studies show that prior exposure of mice to saliva had no detectable effect on numbers of sporozoites delivered by infected mosquitoes, the rate of disappearance of these sporozoites from the skin or infectivity of the sporozoites. Under natural conditions, sporozoites are delivered both to individuals who may exhibit cutaneous hypersensitivity to mosquito bite and to others who may have not yet developed such reactivity. It was tempting to hypothesize that differences in responsiveness to mosquito bite by different individuals might modulate the infectivity of sporozoites delivered into a milieu of changes induced by cutaneous hypersensitivity. Our results with rodent malaria models, however, were unable to support such a hypothesis.
Collapse
|
20
|
Aly ASI, Vaughan AM, Kappe SHI. Malaria parasite development in the mosquito and infection of the mammalian host. Annu Rev Microbiol 2009; 63:195-221. [PMID: 19575563 DOI: 10.1146/annurev.micro.091208.073403] [Citation(s) in RCA: 198] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Plasmodium sporozoites are the product of a complex developmental process in the mosquito vector and are destined to infect the mammalian liver. Attention has been drawn to the mosquito stages and pre-erythrocytic stages owing to recognition that these are bottlenecks in the parasite life cycle and that intervention at these stages can block transmission and prevent infection. Parasite progression in the Anopheles mosquito, sporozoite transmission to the mammalian host by mosquito bite, and subsequent infection of the liver are characterized by extensive migration of invasive stages, cell invasion, and developmental changes. Preparation for the liver phase in the mammalian host begins in the mosquito with an extensive reprogramming of the sporozoite to support efficient infection and survival. Here, we discuss what is known about the molecular and cellular basis of the developmental progression of parasites and their interactions with host tissues in the mosquito and during the early phase of mammalian infection.
Collapse
Affiliation(s)
- Ahmed S I Aly
- Seattle Biomedical Research Institute, Seattle, Washington 98109, USA.
| | | | | |
Collapse
|
21
|
Abstract
Studying the events that occur when a pathogen comes into contact with its host is the basis of the field of infection biology. Over the years, work in this area has revealed many facets of the infection process, including attachment, invasion and colonization by the pathogen, and of the host responses, such as the triggering of the immune system. Recent advancements in imaging technologies, such as multiphoton microscopy (MPM), mean that the field is in the process of taking another big leap forward. MPM allows for cellular-level visualization of the real-time dynamics of infection within the living host. The use of live animal models means that all the interplaying factors of an infection, such as the influences of the immune, lymphatic and vascular systems, can be accounted for. This review outlines the developing field of MPM in pathogen-host interactions, highlighting a number of new insights that have been 'brought to light' using this technique.
Collapse
Affiliation(s)
- Keira Melican
- Swedish Medical Nanoscience Center, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
22
|
Hegge S, Kudryashev M, Smith A, Frischknecht F. Automated classification of Plasmodium sporozoite movement patterns reveals a shift towards productive motility during salivary gland infection. Biotechnol J 2009; 4:903-13. [PMID: 19455538 DOI: 10.1002/biot.200900007] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The invasive stages of malaria and other apicomplexan parasites use a unique motility machinery based on actin, myosin and a number of parasite-specific proteins to invade host cells and tissues. The crucial importance of this motility machinery at several stages of the life cycle of these parasites makes the individual components potential drug targets. The different stages of the malaria parasite exhibit strikingly diverse movement patterns, likely reflecting the varied needs to achieve successful invasion. Here, we describe a Tool for Automated Sporozoite Tracking (ToAST) that allows the rapid simultaneous analysis of several hundred motile Plasmodium sporozoites, the stage of the malaria parasite transmitted by the mosquito. ToAST reliably categorizes different modes of sporozoite movement and can be used for both tracking changes in movement patterns and comparing overall movement parameters, such as average speed or the persistence of sporozoites undergoing a certain type of movement. This allows the comparison of potentially small differences between distinct parasite populations and will enable screening of drug libraries to find inhibitors of sporozoite motility. Using ToAST, we find that isolated sporozoites change their movement patterns towards productive motility during the first week after infection of mosquito salivary glands.
Collapse
Affiliation(s)
- Stephan Hegge
- Department of Parasitology, Hygiene Institute, University of Heidelberg Medical School, Heidelberg, Germany
| | | | | | | |
Collapse
|
23
|
Graewe S, Retzlaff S, Struck N, Janse CJ, Heussler VT. Going live: a comparative analysis of the suitability of the RFP derivatives RedStar, mCherry and tdTomato for intravital and in vitro live imaging of Plasmodium parasites. Biotechnol J 2009; 4:895-902. [PMID: 19492329 DOI: 10.1002/biot.200900035] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Fluorescent proteins have proven to be important tools for in vitro live imaging of parasites and for imaging of parasites within the living host by intravital microscopy. We observed that a red fluorescent transgenic malaria parasite of rodents, Plasmodium berghei-RedStar, is suitable for in vitro live imaging experiments but bleaches rapidly upon illumination in intravital imaging experiments using mice. We have therefore generated two additional transgenic parasite lines expressing the novel red fluorescent proteins tdTomato and mCherry, which have been reported to be much more photostable than first- and second-generation red fluorescent proteins including RedStar. We have compared all three red fluorescent parasite lines for their use in in vitro live and intravital imaging of P. berghei blood and liver parasite stages, using both confocal and wide-field microscopy. While tdTomato bleached almost as rapidly as RedStar, mCherry showed improved photostability and was bright in all experiments performed.
Collapse
Affiliation(s)
- Stefanie Graewe
- Department of Parasitology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | | | | | | |
Collapse
|
24
|
Kinetics of mosquito-injected Plasmodium sporozoites in mice: fewer sporozoites are injected into sporozoite-immunized mice. PLoS Pathog 2009; 5:e1000399. [PMID: 19390607 PMCID: PMC2667259 DOI: 10.1371/journal.ppat.1000399] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2008] [Accepted: 03/26/2009] [Indexed: 11/19/2022] Open
Abstract
Malaria is initiated when the mosquito introduces sporozoites into the skin of a mammalian host. To successfully continue the infection, sporozoites must invade blood vessels in the dermis and be transported to the liver. A significant number of sporozoites, however, may enter lymphatic vessels in the skin or remain in the skin long after the mosquito bite. We have used fluorescence microscopy of Plasmodium berghei sporozoites expressing a fluorescent protein to evaluate the kinetics of sporozoite disappearance from the skin. Sporozoites injected into immunized mice were rapidly immobilized, did not appear to invade dermal blood vessels and became morphologically degraded within several hours. Strikingly, mosquitoes introduced significantly fewer sporozoites into immunized than into non-immunized mice, presumably by formation of an immune complex between soluble sporozoite antigens in the mosquito saliva and homologous host antibodies at the proboscis tip. These results indicate that protective antibodies directed against sporozoites may function both by reducing the numbers of sporozoites injected into immunized hosts and by inhibiting the movement of injected sporozoites into dermal blood vessels. Malaria is initiated by a mosquito injecting malaria sporozoites into the skin. To successfully continue the infection, sporozoites must then invade blood vessels in skin for transportation to the liver. However, the majority of these injected sporozoites are unable to reach the blood. The numbers of sporozoites that successfully invade the blood may influence the characteristics of the subsequent clinical malaria infection. We studied this by microscopy with fluorescent sporozoites of the rodent malaria parasite Plasmodium berghei injected into mice by mosquitoes. Sporozoites introduced into mice that have been immunized against sporozoites become immobilized and cannot reach the blood; those that remain at the bite site become degraded within several hours. Strikingly, mosquitoes introduce significantly fewer sporozoites into skin of immunized mice. These findings indicate that antibodies directed against sporozoites seem to function both by reducing the numbers of sporozoites injected into immunized hosts in the first place and then by inhibiting the movement of the injected sporozoites into the bloodstream.
Collapse
|
25
|
Frevert U, Usynin I, Baer K, Klotz C. Plasmodium sporozoite passage across the sinusoidal cell layer. Subcell Biochem 2008; 47:182-97. [PMID: 18512352 DOI: 10.1007/978-0-387-78267-6_15] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Malaria sporozoites must cross at least two cell barriers to reach their initial site of replication in the mammalian host. After transmission into the skin by an infected mosquito, they migrate towards small dermal capillaries, traverse the vascular endothelial layer, and rapidly home to the liver. To infect hepatocytes, the parasites must cross the sinusoidal cell layer, composed of specialized highly fenestrated sinusoidal endothelia and Kupffer cells, the resident macrophages of the liver (Fig. 1). The exact route Plasmodium sporozoites take to hepatocytes has been subject of controversial discussions for many years. Recent cell biological, microscopic, and genetic approaches have considerably enhanced our understanding of the initial events leading to the establishment of a malaria infection in the liver.
Collapse
Affiliation(s)
- Ute Frevert
- NYU School of Medicine, Department of Medical Parasitology, 341 E. 25 Street, New York, New York 10010, USA.
| | | | | | | |
Collapse
|
26
|
Santos JM, Lebrun M, Daher W, Soldati D, Dubremetz JF. Apicomplexan cytoskeleton and motors: key regulators in morphogenesis, cell division, transport and motility. Int J Parasitol 2008; 39:153-62. [PMID: 19028497 DOI: 10.1016/j.ijpara.2008.10.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2008] [Revised: 10/13/2008] [Accepted: 10/16/2008] [Indexed: 10/21/2022]
Abstract
Protozoan parasites of the phylum Apicomplexa undergo a lytic cycle whereby a single zoite produced by the previous cycle has to encounter a host cell, invade it, multiply to differentiate into a new zoite generation and escape to resume a new cycle. At every step of this lytic cycle, the cytoskeleton and/or the gliding motility apparatus play a crucial role and recent results have elucidated aspects of these processes, especially in terms of the molecular characterization and interaction of the increasing number of partners involved, and the signalling mechanisms implicated. The present review aims to summarize the most recent findings in the field.
Collapse
Affiliation(s)
- Joana M Santos
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva CMU, 1 rue Michel-Servet, 1211 Geneva 4, Switzerland
| | | | | | | | | |
Collapse
|
27
|
Vaughan AM, Aly ASI, Kappe SHI. Malaria parasite pre-erythrocytic stage infection: gliding and hiding. Cell Host Microbe 2008; 4:209-18. [PMID: 18779047 DOI: 10.1016/j.chom.2008.08.010] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Accepted: 08/20/2008] [Indexed: 12/24/2022]
Abstract
In malaria, the red blood cell-infectious form of the Plasmodium parasite causes illness and the possible death of infected hosts. The initial infection in the liver caused by the mosquito-borne sporozoite parasite stage, however, causes little pathology and no symptoms. Nevertheless, pre-erythrocytic parasite stages are attracting passionate research efforts not least because they are the most promising targets for malaria vaccine development. Here, we review how the infectious sporozoite makes its way to the liver and subsequently develops within hepatocytes. We discuss the factors, both parasite and host, involved in the interactions that occur during this "silent" phase of infection.
Collapse
|
28
|
Frevert U, Moreno A, Calvo-Calle JM, Klotz C, Nardin E. Imaging effector functions of human cytotoxic CD4+ T cells specific for Plasmodium falciparum circumsporozoite protein. Int J Parasitol 2008; 39:119-32. [PMID: 18723023 DOI: 10.1016/j.ijpara.2008.06.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Accepted: 06/09/2008] [Indexed: 10/21/2022]
Abstract
Malaria vaccines, comprised of irradiated Plasmodium falciparum sporozoites or a synthetic peptide containing T and B cell epitopes of the circumsporozoite protein (CSP), elicit multifunctional cytotoxic and non-cytotoxic CD4(+) T cells in immunised volunteers. Both lytic and non-lytic CD4(+)T cell clones recognised a series of overlapping epitopes within a 'universal' T cell epitope EYLNKIQNSLSTEWSPCSVT of CSP (NF54 isolate) that was presented in the context of multiple DR molecules. Lytic activity directly correlated with T cell receptor (TCR) functional avidity as measured by stimulation indices and recognition of naturally occurring variant peptides. CD4(+) T cell-mediated cytotoxicity was contact-dependent and did not require de novo synthesis of cytotoxic mediators, suggesting a granule-mediated mechanism. Live cell imaging of the interaction of effector and target cells demonstrated that CD4(+) cytotoxic T cells recognise target cells with their leading edge, reorient their cytotoxic granules towards the zone of contact, and form a stable immunological synapse. CTL attacks induced chromatin condensation, nuclear fragmentation and formation of apoptotic bodies in target cells. Together, these findings suggest that CD4(+) CTLs trigger target cell apoptosis via classical perforin/granzyme-mediated cytotoxicity, similar to CD8(+) CTLs, and these multifunctional sporozoite- and peptide-induced CD4(+) T cells have the potential to play a direct role as effector cells in targeting the exoerythrocytic forms within the liver.
Collapse
Affiliation(s)
- Ute Frevert
- Department of Medical Parasitology, New York University School of Medicine, 341 E 25 St, New York, NY 10010, USA
| | | | | | | | | |
Collapse
|
29
|
Biotech meetings. Biotechnol J 2008. [DOI: 10.1002/biot.200890073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
30
|
Schmidt-Christensen A, Sturm A, Horstmann S, Heussler VT. Expression and processing of Plasmodium berghei SERA3 during liver stages. Cell Microbiol 2008; 10:1723-34. [PMID: 18419771 PMCID: PMC2613260 DOI: 10.1111/j.1462-5822.2008.01162.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Cysteine proteases mediate liberation of Plasmodium berghei merozoites from infected hepatocytes. In an attempt to identify the responsible parasite proteases, we screened the genome of P. berghei for cysteine protease-encoding genes. RT-PCR analyses revealed that transcription of four out of five P. berghei serine repeat antigen (PbSERA) genes was strongly upregulated in late liver stages briefly before the parasitophorous vacuole membrane ruptured to release merozoites into the host cell cytoplasm, suggesting a role of PbSERA proteases in these processes. In order to characterize PbSERA3 processing, we raised an antiserum against a non-conserved region of the protein and generated a transgenic P. berghei strain expressing a TAP-tagged PbSERA3 under the control of the endogenous promoter. Immunofluorescence assays revealed that PbSERA3 leaks into the host cell cytoplasm during merozoite development, where it might contribute to host cell death or activate host cell proteases that execute cell death. Importantly, processed PbSERA3 has been detected by Western blot analysis in cell extracts of schizont-infected cells and merozoite-infected detached hepatic cells.
Collapse
|
31
|
Chen Q, Wang H. Implications of imaging malaria sporozoites. Trends Parasitol 2008; 24:106-9. [PMID: 18280209 DOI: 10.1016/j.pt.2007.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2007] [Revised: 12/11/2007] [Accepted: 11/15/2007] [Indexed: 11/26/2022]
Abstract
The sporozoites of Plasmodium parasites undergo several transmigrations before their establishment in the hepatocytes of a vertebrate host. Techniques that illustrate parasite intra-vital migration and their interaction with host cells will advance the understanding of parasite biology. In a recent publication, Amino et al. provided a detailed protocol for in vivo imaging of Plasmodium berghei sporozoites in the dermis. The report has important implications in the dissection of malaria parasite biology.
Collapse
Affiliation(s)
- Qijun Chen
- Key Laboratory of Zoonosis, Ministry of Education, Changchun 130062, China. [corrected]
| | | |
Collapse
|
32
|
Korn CB, Schwarz US. Mean first passage times for bond formation for a Brownian particle in linear shear flow above a wall. J Chem Phys 2007; 126:095103. [PMID: 17362131 DOI: 10.1063/1.2464080] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Motivated by cell adhesion in hydrodynamic flow, here the authors study bond formation between a spherical Brownian particle in linear shear flow carrying receptors for ligands covering the boundary wall. They derive the appropriate Langevin equation which includes multiplicative noise due to position-dependent mobility functions resulting from the Stokes equation. They present a numerical scheme which allows to simulate it with high accuracy for all model parameters, including shear rate and three parameters describing receptor geometry (distance, size, and height of the receptor patches). In the case of homogeneous coating, the mean first passage time problem can be solved exactly. In the case of position-resolved receptor-ligand binding, they identify different scaling regimes and discuss their biological relevance.
Collapse
Affiliation(s)
- C B Korn
- University of Heidelberg, Im Neuenheimer Feld 293, D-69120, Germany
| | | |
Collapse
|
33
|
Janse CJ, Franke-Fayard B, Waters AP. Selection by flow-sorting of genetically transformed, GFP-expressing blood stages of the rodent malaria parasite, Plasmodium berghei. Nat Protoc 2007; 1:614-23. [PMID: 17406288 DOI: 10.1038/nprot.2006.88] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This protocol describes a methodology for the genetic transformation of the rodent malaria parasite Plasmodium berghei and the subsequent selection of transformed parasites expressing green fluorescent protein (GFP) by flow-sorting. It provides methods for: transfection of the schizont stage with DNA constructs that contain gfp as the selectable marker; selection of fluorescent mutants by flow-sorting; and injection of flow-sorted, GFP-expressing parasites into mice and the subsequent collection of transformed parasites. The use of two different promoters for the expression of GFP is described; these two promoters require slightly different procedures for the selection of mutants. The protocol enables the collection of transformed parasites within 10-12 days after transfection. The genetic modification of P. berghei is widely used to investigate gene function in Plasmodium sp. The application of flow-sorting to the selection of transformed parasites increases the possibilities of parasite mutagenesis, by effectively expanding the range of selectable markers.
Collapse
|
34
|
Franke-Fayard B, Waters AP, Janse CJ. Real-time in vivo imaging of transgenic bioluminescent blood stages of rodent malaria parasites in mice. Nat Protoc 2007; 1:476-85. [PMID: 17406270 DOI: 10.1038/nprot.2006.69] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This protocol describes a methodology for imaging the sequestration of infected erythrocytes of the rodent malaria parasite Plasmodium berghei in the bodies of live mice or in dissected organs, using a transgenic parasite that expresses luciferase. Real-time imaging of infected erythrocytes is performed by measuring bioluminescence produced by the enzymatic reaction between luciferase and its substrate luciferin, which is injected into the mice several minutes prior to imaging. The bioluminescence signal is detected by an intensified charge-coupled device (I-CCD) photon-counting video camera. Sequestration of infected erythrocytes is imaged during short-term infections with synchronous parasite development or during ongoing infections. With this technology, sequestration patterns of the schizont stage can be quantitatively analyzed within 1-2 d after infection. Real-time in vivo imaging of infected erythrocytes will provide increased insights into the dynamics of sequestration and its role in pathology, and can be used to evaluate strategies that prevent sequestration.
Collapse
Affiliation(s)
- Blandine Franke-Fayard
- Malaria Research Group, Department of Parasitology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | | | | |
Collapse
|
35
|
Tilley L, McFadden G, Cowman A, Klonis N. Illuminating Plasmodium falciparum-infected red blood cells. Trends Parasitol 2007; 23:268-77. [PMID: 17434344 DOI: 10.1016/j.pt.2007.04.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2006] [Revised: 03/21/2007] [Accepted: 04/04/2007] [Indexed: 11/20/2022]
Abstract
The malaria parasite undergoes a remarkable series of morphological transformations, which underpin its life in both human and mosquito hosts. The advent of molecular transfection technology coupled with the ability to introduce fluorescent reporter proteins that faithfully track and expose the activities of parasite proteins has revolutionized our view of parasite cell biology. The greatest insights have been realized in the erythrocyte stages of Plasmodium falciparum. P. falciparum invades and remodels the human erythrocyte: it feeds on haemoglobin, grows and divides, and subverts the physiology of its hapless host. Fluorescent proteins have been employed to track and dissect each of these processes and have revealed details and exposed new paradigms.
Collapse
Affiliation(s)
- Leann Tilley
- Department of Biochemistry, La Trobe University, Melbourne, Victoria 3086, Australia.
| | | | | | | |
Collapse
|
36
|
Cyrklaff M, Kudryashev M, Leis A, Leonard K, Baumeister W, Menard R, Meissner M, Frischknecht F. Cryoelectron tomography reveals periodic material at the inner side of subpellicular microtubules in apicomplexan parasites. ACTA ACUST UNITED AC 2007; 204:1281-7. [PMID: 17562819 PMCID: PMC2118598 DOI: 10.1084/jem.20062405] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Microtubules are dynamic cytoskeletal structures important for cell division, polarity, and motility and are therefore major targets for anticancer and antiparasite drugs. In the invasive forms of apicomplexan parasites, which are highly polarized and often motile cells, exceptionally stable subpellicular microtubules determine the shape of the parasite, and serve as tracks for vesicle transport. We used cryoelectron tomography to image cytoplasmic structures in three dimensions within intact, rapidly frozen Plasmodium sporozoites. This approach revealed microtubule walls that are extended at the luminal side by an additional 3 nm compared to microtubules of mammalian cells. Fourier analysis revealed an 8-nm longitudinal periodicity of the luminal constituent, suggesting the presence of a molecule interacting with tubulin dimers. In silico generation and analysis of microtubule models confirmed this unexpected topology. Microtubules from extracted sporozoites and Toxoplasma gondii tachyzoites showed a similar density distribution, suggesting that the putative protein is conserved among Apicomplexa and serves to stabilize microtubules.
Collapse
Affiliation(s)
- Marek Cyrklaff
- Department of Molecular Structural Biology, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Sinnis P, Coppi A. A long and winding road: the Plasmodium sporozoite's journey in the mammalian host. Parasitol Int 2007; 56:171-8. [PMID: 17513164 PMCID: PMC1995443 DOI: 10.1016/j.parint.2007.04.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2007] [Accepted: 04/17/2007] [Indexed: 11/21/2022]
Abstract
The Plasmodium sporozoite, the infectious stage of the malaria parasite, makes a remarkable journey in its mammalian host. Here we review our current knowledge of the molecular and cellular basis of this journey, which begins in the skin and ends in the hepatocyte.
Collapse
Affiliation(s)
- Photini Sinnis
- Department of Medical Parasitology, New York University School of Medicine, 341 East 25th Street, New York, NY 10010, United States.
| | | |
Collapse
|
38
|
Meissner M, Agop-Nersesian C, Sullivan WJ. Molecular tools for analysis of gene function in parasitic microorganisms. Appl Microbiol Biotechnol 2007; 75:963-75. [PMID: 17401559 DOI: 10.1007/s00253-007-0946-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2007] [Revised: 03/12/2007] [Accepted: 03/12/2007] [Indexed: 01/01/2023]
Abstract
With the completion of several genome sequences for parasitic protozoa, research in molecular parasitology entered the "post-genomic" era. Accompanied by global transcriptome and proteome analysis, huge datasets have been generated that have added many novel candidates to the list of drug and vaccine targets. The challenge is now to validate these factors and to bring science back to the bench to perform a detailed characterization. In some parasites, like Trypanosoma brucei, high-throughput genetic screens have been established using RNA interference [for a detailed review, see Motyka and Englund (2004)]. In most protozoan parasites, however, more time-consuming approaches have to be employed to identify and characterize the function of promising candidates in detail. This review aims to summarize the status of molecular genetic tools available for a variety of protozoan pathogens and discuss how they can be implemented to advance our understanding of parasite biology.
Collapse
Affiliation(s)
- Markus Meissner
- Hygieneinstitut, Abteilung Parasitologie, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany.
| | | | | |
Collapse
|
39
|
Frischknecht F, Amino R, Franke-Fayard B, Janse C, Waters A, Ménard R. Imaging Parasites in Vivo. IMAGING CELLULAR AND MOLECULAR BIOLOGICAL FUNCTIONS 2007. [DOI: 10.1007/978-3-540-71331-9_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
40
|
Baker RP, Wijetilaka R, Urban S. Two Plasmodium rhomboid proteases preferentially cleave different adhesins implicated in all invasive stages of malaria. PLoS Pathog 2006; 2:e113. [PMID: 17040128 PMCID: PMC1599764 DOI: 10.1371/journal.ppat.0020113] [Citation(s) in RCA: 176] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2006] [Accepted: 09/14/2006] [Indexed: 11/18/2022] Open
Abstract
Invasion of host cells by the malaria pathogen Plasmodium relies on parasite transmembrane adhesins that engage host-cell receptors. Adhesins must be released by cleavage before the parasite can enter the cell, but the processing enzymes have remained elusive. Recent work indicates that the Toxoplasma rhomboid intramembrane protease TgROM5 catalyzes this essential cleavage. However, Plasmodium does not encode a direct TgROM5 homolog. We examined processing of the 14 Plasmodium falciparum adhesins currently thought to be involved in invasion by both model and Plasmodium rhomboid proteases in a heterologous assay. While most adhesins contain aromatic transmembrane residues and could not be cleaved by nonparasite rhomboid proteins, including Drosophila Rhomboid-1, Plasmodium falciparum rhomboid protein (PfROM)4 (PFE0340c) was able to process these adhesins efficiently and displayed novel substrate specificity. Conversely, PfROM1 (PF11_0150) shared specificity with rhomboid proteases from other organisms and was the only PfROM able to cleave apical membrane antigen 1 (AMA1). PfROM 1 and/or 4 was thus able to cleave diverse adhesins including TRAP, CTRP, MTRAP, PFF0800c, EBA-175, BAEBL, JESEBL, MAEBL, AMA1, Rh1, Rh2a, Rh2b, and Rh4, but not PTRAMP, and cleavage relied on the adhesin transmembrane domains. Swapping transmembrane regions between BAEBL and AMA1 switched the relative preferences of PfROMs 1 and 4 for these two substrates. Our analysis indicates that PfROMs 1 and 4 function with different substrate specificities that together constitute the specificity of TgROM5 to cleave diverse adhesins. This is the first enzymatic analysis of Plasmodium rhomboid proteases and suggests an involvement of PfROMs in all invasive stages of the malaria lifecycle, in both the vertebrate host and the mosquito vector. Malaria is a devastating global disease that afflicts over 10% of the world's population, claiming between 1 and 3 million lives annually. Invasion of host cells by the malaria parasite Plasmodium ultimately requires enzymes to release close contacts made between the parasite and host cell, but these enzymes have not been identified. Rhomboid enzymes were previously found to be involved in this process in the related pathogen Toxoplasma. The present work examined the activity of Plasmodium rhomboid enzymes, and revealed that two Plasmodium rhomboid enzymes can cleave most, if not all, of the proteins currently known to mediate contacts between the parasite and host-cell membranes during invasion. The two rhomboid enzymes had different specificities for the different target proteins, but together could process all of the proteins that the similar Toxoplasma rhomboid enzyme could process alone. This analysis suggests that rhomboid enzymes may be essential for the ability of the parasite to invade host cells through different pathways both in the human and mosquito hosts, and therefore offers a possible new therapeutic target to explore for treating or controlling the devastating effects of malaria.
Collapse
Affiliation(s)
- Rosanna P Baker
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Center of Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Ruvini Wijetilaka
- Center of Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Sinisa Urban
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Center of Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
41
|
Tarun AS, Baer K, Dumpit RF, Gray S, Lejarcegui N, Frevert U, Kappe SHI. Quantitative isolation and in vivo imaging of malaria parasite liver stages. Int J Parasitol 2006; 36:1283-93. [PMID: 16890231 DOI: 10.1016/j.ijpara.2006.06.009] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2006] [Revised: 06/05/2006] [Accepted: 06/09/2006] [Indexed: 11/24/2022]
Abstract
The liver stages of Plasmodium, the causative agent of malaria, are the least explored forms in the parasite's life cycle despite their recognition as key vaccine and drug targets. In vivo experimental access to liver stages of human malaria parasites is practically prohibited and therefore rodent model malaria parasites have been used for in vivo studies. However, even in rodent models progress in the analysis of liver stages has been limited, mainly due to their low abundance and associated difficulties in visualisation and isolation. Here, we present green fluorescent protein (GFP)-tagged Plasmodium yoelii rodent malaria parasite liver infections in BALB/c mice as an excellent quantitative model for the live visualisation and isolation of the so far elusive liver stages. We believe P. yoelii GFP-tagged liver stages allow, for the first time, the efficient quantitative isolation of intact early and late liver stage-infected hepatocyte units by fluorescence activated cell sorting. GFP-tagged liver stages are also well suited for intravital imaging, allowing us for the first time to visualise them in real time. We identify previously unrecognised features of liver stages including vigorous parasite movement and expulsion of 'extrusomes'. Intravital imaging thus reveals new, important information on the malaria parasite's transition from tissue to blood stage.
Collapse
Affiliation(s)
- Alice S Tarun
- Seattle Biomedical Research Institute, Seattle, WA 98109, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Over the millennia, pathogens have coevolved with their hosts and acquired the ability to intercept, disrupt, mimic, and usurp numerous signaling pathways of those hosts. The study of host/pathogen interactions thus not only teaches us about the intricate biology of these parasitic invaders but also provides interesting insights into basic cellular processes both at the level of the individual cell and more globally throughout the organism. Host/pathogen relationships also provide insights into the evolutionary forces that shape biological diversity. Here we review a few recent examples of how viruses, bacteria, and parasites manipulate tyrosine kinase-mediated and Rho guanosine triphosphatase-mediated signaling pathways of their hosts to achieve efficient entry, replication, and exit during their infectious cycles.
Collapse
Affiliation(s)
- Sylvia Münter
- Department of Parasitology, Hygiene Institute, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | | | | |
Collapse
|
43
|
Frischknecht F, Martin B, Thiery I, Bourgouin C, Menard R. Using green fluorescent malaria parasites to screen for permissive vector mosquitoes. Malar J 2006; 5:23. [PMID: 16569221 PMCID: PMC1450296 DOI: 10.1186/1475-2875-5-23] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2005] [Accepted: 03/28/2006] [Indexed: 11/10/2022] Open
Abstract
Background The Plasmodium species that infect rodents, particularly Plasmodium berghei and Plasmodium yoelii, are useful to investigate host-parasite interactions. The mosquito species that act as vectors of human plasmodia in South East Asia, Africa and South America show different susceptibilities to infection by rodent Plasmodium species. P. berghei and P. yoelii infect both Anopheles gambiae and Anopheles stephensi, which are found mainly in Africa and Asia, respectively. However, it was reported that P. yoelii can infect the South American mosquito, Anopheles albimanus, while P. berghei cannot. Methods P. berghei lines that express the green fluorescent protein were used to screen for mosquitoes that are susceptible to infection by P. berghei. Live mosquitoes were examined and screened for the presence of a fluorescent signal in the abdomen. Infected mosquitoes were then examined by time-lapse microscopy to reveal the dynamic behaviour of sporozoites in haemolymph and extracted salivary glands. Results A single fluorescent oocyst can be detected in live mosquitoes and P. berghei can infect A. albimanus. As in other mosquitoes, P. berghei sporozoites can float through the haemolymph and invade A. albimanus salivary glands and they are infectious in mice after subcutaneous injection. Conclusion Fluorescent Plasmodium parasites can be used to rapidly screen susceptible mosquitoes. These results open the way to develop a laboratory model in countries where importation of A. gambiae and A. stephensi is not allowed.
Collapse
Affiliation(s)
- Friedrich Frischknecht
- Unité de Biologie et Génétique du Paludisme, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
- Department of Parasitology, University of Heidelberg Medical School, Tel 49-6221-566537, Fax 49-6221-564643, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Beatrice Martin
- Unité de Biologie et Génétique du Paludisme, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
- CNRS, Institut Alfred Fessard, Neurobiologie Génétique et Intégrative, Avenue de la Terrasse, 91198 Gif-sur-Yvette, France
| | - Isabelle Thiery
- Unité de Biologie et Génétique du Paludisme, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
- Centre de Production et d'Infection des Anopheles, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Catherine Bourgouin
- Unité de Biologie et Génétique du Paludisme, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
- Centre de Production et d'Infection des Anopheles, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Robert Menard
- Unité de Biologie et Génétique du Paludisme, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
| |
Collapse
|
44
|
Heussler V, Doerig C. In vivo imaging enters parasitology. Trends Parasitol 2006; 22:192-5; discussion 195-6. [PMID: 16545613 DOI: 10.1016/j.pt.2006.03.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Revised: 02/07/2006] [Accepted: 03/02/2006] [Indexed: 11/19/2022]
Abstract
In vivo infection routes of parasites have remained something of a "black box", in which only snapshot views of fixed tissues are available. Clearly, there exists a strong need for imaging approaches to visualise living parasites within intact organs and animals. In vivo imaging of fluorescent Plasmodium parasites now provides us with exciting insights into the infection process, from the bite of the infected mosquito to the invasion of liver cells, and alternative approaches using luciferase-expressing parasites have been used to monitor their dissemination in mice. This rapidly developing field will go a long way towards deepening our understanding of host-parasite interactions at different levels.
Collapse
Affiliation(s)
- Volker Heussler
- Bernhard Nocht Institute for Tropical Medicine, Department of Molecular Parasitology, Malaria laboratory I, Bernhard-Nocht-Strasse 74, 20359 Hamburg, Germany.
| | | |
Collapse
|
45
|
Lehmann MJ, Frischknecht F. Surfing Through a Sea of Sharks: Report on the British Society for Cell Biology Meeting on ‘Signaling and Cytoskeletal Dynamics During Infection’, October 2-5, 2005, Edinburgh, Scotland. Traffic 2006; 7:479-87. [PMID: 16536746 DOI: 10.1111/j.1600-0854.2006.00402.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Maik J Lehmann
- Department of Virology, Hygiene Institute, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | | |
Collapse
|
46
|
Heussler V, Sturm A, Langsley G. Regulation of host cell survival by intracellular Plasmodium and Theileria parasites. Parasitology 2006; 132 Suppl:S49-60. [PMID: 17018165 DOI: 10.1017/s0031182006000850] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Plasmodium and Theileria parasites are obligate intracellular protozoa of the phylum Apicomplexa. Theileria infection of bovine leukocytes induces transformation of host cells and infected leukocytes can be kept indefinitely in culture. Theileria-dependent host cell transformation has been the subject of interest for many years and the molecular basis of this unique phenomenon is quite well understood. The equivalent life cycle stage of Plasmodium is the infection of mammalian hepatocytes, where parasites reside for 2-7 days depending on the species. Some of the molecular details of parasite-host interactions in P. berghei-infected hepatocytes have emerged only very recently. Similar to what has been shown for Theileria-infected leukocytes these data suggest that malaria parasites within hepatocytes also protect their host cell from programmed cell death. However, the strategies employed to inhibit host cell apoptotic pathways appear to be different to those used by Theileria. This review discusses similarities and differences at the molecular level of Plasmodium- and Theileria-induced regulation of the host cell survival machinery.
Collapse
Affiliation(s)
- V Heussler
- Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Str. 74, 20359 Hamburg, Germany.
| | | | | |
Collapse
|
47
|
Engelmann S, Sinnis P, Matuschewski K. Transgenic Plasmodium berghei sporozoites expressing beta-galactosidase for quantification of sporozoite transmission. Mol Biochem Parasitol 2005; 146:30-7. [PMID: 16316690 PMCID: PMC1853313 DOI: 10.1016/j.molbiopara.2005.10.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2005] [Revised: 10/12/2005] [Accepted: 10/19/2005] [Indexed: 11/20/2022]
Abstract
Malaria transmission occurs during a blood-meal of an infected Anopheles mosquito. Visualization and quantification of sporozoites along the journey from the mosquito midgut, where they develop, to the vertebrate liver, their final target organ, is important for understanding many aspects of sporozoite biology. Here we describe the generation of Plasmodium berghei parasites that express the reporter gene lacZ as a stable transgene, under the control of the sporozoite-specific CSP promoter. Transgenic sporozoites expressing beta-galactosidase can be simply visualized and quantified in an enzymatic assay. In addition, these sporozoites can be used to quantify sporozoites deposited in subcutaneous tissue during natural infection.
Collapse
Affiliation(s)
- Sabine Engelmann
- Department of Parasitology, Heidelberg University School of Medicine, INF 324, 69120 Heidelberg, Germany
| | - Photini Sinnis
- Department of Medical and Molecular Parasitology, New York University School of Medicine, 341 E. 25th St., New York, NY 10010, USA
- * Corresponding author. Tel.: +1 212 263 6818; fax: +1 212 263 8116
| | - Kai Matuschewski
- Department of Parasitology, Heidelberg University School of Medicine, INF 324, 69120 Heidelberg, Germany
- ** Corresponding author. Tel.: +49 6221 568284; fax: +49 6221 564643
| |
Collapse
|