1
|
Pražák V, Vasishtan D, Grünewald K, Douglas RG, Ferreira JL. Molecular architecture of glideosome and nuclear F-actin in Plasmodium falciparum. EMBO Rep 2025; 26:1984-1996. [PMID: 40128412 PMCID: PMC12019134 DOI: 10.1038/s44319-025-00415-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/30/2025] [Accepted: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
Actin-based motility is required for the transmission of malaria sporozoites. While this has been shown biochemically, filamentous actin has remained elusive and has not been directly visualised inside the parasite. Using focused ion beam milling and electron cryo-tomography, we studied dynamic actin filaments in unperturbed Plasmodium falciparum cells for the first time. This allowed us to dissect the assembly, path and fate of actin filaments during parasite gliding and determine a complete 3D model of F-actin within sporozoites. We observe micrometre long actin filaments, much longer than expected from in vitro studies. After their assembly at the parasite's apical end, actin filaments continue to grow as they are transported down the cell as part of the glideosome machinery, and are disassembled at the basal end in a rate-limiting step. Large pores in the IMC, constrained to the basal end, may facilitate actin exchange between the pellicular space and cytosol for recycling and maintenance of directional flow. The data also reveal striking actin bundles in the nucleus. Implications for motility and transmission are discussed.
Collapse
Affiliation(s)
- Vojtěch Pražák
- Leibniz-Institut für Virologie (LIV), Hamburg, 20251, Germany
- Centre for Structural Systems Biology, Hamburg, 22607, Germany
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Daven Vasishtan
- Leibniz-Institut für Virologie (LIV), Hamburg, 20251, Germany
- Centre for Structural Systems Biology, Hamburg, 22607, Germany
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Kay Grünewald
- Leibniz-Institut für Virologie (LIV), Hamburg, 20251, Germany
- Centre for Structural Systems Biology, Hamburg, 22607, Germany
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
- Department of Chemistry, Universität Hamburg, Hamburg, 20148, Germany
| | - Ross G Douglas
- Biochemistry and Molecular Biology, Interdisciplinary Research Centre and Molecular Infection Biology, Biomedical Research Centre Seltersberg, Justus Liebig University Giessen, Giessen, 35392, Germany
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, 35392, Germany
| | - Josie L Ferreira
- Institute for Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
2
|
Frangos SM, Damrich S, Gueiber D, Sanchez CP, Wiedemann P, Schwarz US, Hamprecht FA, Lanzer M. Deep learning image analysis for continuous single-cell imaging of dynamic processes in Plasmodium falciparum-infected erythrocytes. Commun Biol 2025; 8:487. [PMID: 40133663 PMCID: PMC11937545 DOI: 10.1038/s42003-025-07894-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
Continuous high-resolution imaging of the disease-mediating blood stages of the human malaria parasite Plasmodium falciparum faces challenges due to photosensitivity, small parasite size, and the anisotropy and large refractive index of host erythrocytes. Previous studies often relied on snapshot galleries from multiple cells, limiting the investigation of dynamic cellular processes. We present a workflow enabling continuous, single-cell monitoring of live parasites throughout the 48-hour intraerythrocytic life cycle with high spatial and temporal resolution. This approach integrates label-free, three-dimensional differential interference contrast and fluorescence imaging using an Airyscan microscope, automated cell segmentation through pre-trained deep-learning algorithms, and 3D rendering for visualization and time-resolved analyses. As a proof of concept, we applied this workflow to study knob-associated histidine-rich protein (KAHRP) export into the erythrocyte compartment and its clustering beneath the plasma membrane. Our methodology opens avenues for in-depth exploration of dynamic cellular processes in malaria parasites, providing a valuable tool for further investigations.
Collapse
Affiliation(s)
- Sophia M Frangos
- Heidelberg University, Medical Faculty, University Hospital Heidelberg, Center for Infectious Diseases, Parasitology, Im Neuenheimer Feld 324, Heidelberg, Germany
| | - Sebastian Damrich
- Heidelberg University, Interdisciplinary Center for Scientific Computing (IWR), Im Neuenheimer Feld 205, Heidelberg, Germany
- Hertie Institute for AI in Brain Health, University of Tübingen, Otfried-Müller-Straße 25, Tübingen, Germany
| | - Daniele Gueiber
- Heidelberg University, Medical Faculty, University Hospital Heidelberg, Center for Infectious Diseases, Parasitology, Im Neuenheimer Feld 324, Heidelberg, Germany
- University of Applied Sciences Mannheim, Institute of Molecular and Cell Biology, Paul-Wittsack-Strasse 10, Mannheim, Germany
| | - Cecilia P Sanchez
- Heidelberg University, Medical Faculty, University Hospital Heidelberg, Center for Infectious Diseases, Parasitology, Im Neuenheimer Feld 324, Heidelberg, Germany
| | - Philipp Wiedemann
- University of Applied Sciences Mannheim, Institute of Molecular and Cell Biology, Paul-Wittsack-Strasse 10, Mannheim, Germany
| | - Ulrich S Schwarz
- Heidelberg University, BioQuant and Institute for Theoretical Physics, Philosophenweg 19, Heidelberg, Germany
| | - Fred A Hamprecht
- Heidelberg University, Interdisciplinary Center for Scientific Computing (IWR), Im Neuenheimer Feld 205, Heidelberg, Germany
| | - Michael Lanzer
- Heidelberg University, Medical Faculty, University Hospital Heidelberg, Center for Infectious Diseases, Parasitology, Im Neuenheimer Feld 324, Heidelberg, Germany.
| |
Collapse
|
3
|
Cabral TB, de Oliveira AC, de Melo GC, Rodrigues-Soares F. Pharmacogenetics of the Treatment of Neglected Diseases. Genes (Basel) 2025; 16:54. [PMID: 39858601 PMCID: PMC11764765 DOI: 10.3390/genes16010054] [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: 11/30/2024] [Revised: 12/20/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Pharmacogenetics (PGx) aims to identify individuals more likely to suffer from adverse reactions or therapeutic failure in drug treatments. However, despite most of the evidence in this area being from European populations, some diseases have also been neglected, such as HIV infection, malaria, and tuberculosis. With this review, we aim to emphasize which pharmacogenetic tests are ready to be implemented in treating neglected diseases that have some evidence and call attention to what is missing for these three diseases. METHODS A critical literature review on the PGx of HIV infection, malaria, and tuberculosis was performed. RESULTS There are three PGx guidelines for antiretroviral drugs used in HIV infection, one for malaria, and none for tuberculosis. Some evidence is already available, and some genes have already been identified, such as CYP2D6 for primaquine treatment and NAT2 for isoniazid. However, some barriers to the implementation are the lack of evidence due to the few studies on the diseases themselves and the admixture of the most affected populations, which must be considered, given the genetic differentiation of these populations. CONCLUSIONS PGx tests such as abacavir are already implemented in some places, and efavirenz/atazanavir is ready to implement if this medication is used. Other gene-drug associations were found but still do not present a clear recommendation. We call attention to the need to generate more evidence for testing treatments for other neglected diseases, such as malaria and tuberculosis, given their epidemiological importance and for the public health of less favored populations.
Collapse
Affiliation(s)
- Tiffany Borges Cabral
- Department of Pathology, Genetics and Evolution, Federal University of Triângulo Mineiro, Uberaba 38025-180, Brazil;
| | - Amanda Carvalho de Oliveira
- Postgraduate Program in Tropical Medicine, Amazonas State University (PPGMT/UEA), Manaus 69040-000, Brazil; (A.C.d.O.); (G.C.d.M.)
- Tropical Medicine Foundation (FMT-HVD) Dr. Heitor Vieira Dourado, Manaus 69040-000, Brazil
| | - Gisely Cardoso de Melo
- Postgraduate Program in Tropical Medicine, Amazonas State University (PPGMT/UEA), Manaus 69040-000, Brazil; (A.C.d.O.); (G.C.d.M.)
- Tropical Medicine Foundation (FMT-HVD) Dr. Heitor Vieira Dourado, Manaus 69040-000, Brazil
| | - Fernanda Rodrigues-Soares
- Department of Pathology, Genetics and Evolution, Federal University of Triângulo Mineiro, Uberaba 38025-180, Brazil;
| |
Collapse
|
4
|
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
|
5
|
Otesteanu CF, Caldelari R, Heussler V, Sznitman R. Machine learning for predicting Plasmodium liver stage development in vitro using microscopy imaging. Comput Struct Biotechnol J 2024; 24:334-342. [PMID: 38690550 PMCID: PMC11059334 DOI: 10.1016/j.csbj.2024.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 05/02/2024] Open
Abstract
Malaria, a significant global health challenge, is caused by Plasmodium parasites. The Plasmodium liver stage plays a pivotal role in the establishment of the infection. This study focuses on the liver stage development of the model organism Plasmodium berghei, employing fluorescent microscopy imaging and convolutional neural networks (CNNs) for analysis. Convolutional neural networks have been recently proposed as a viable option for tasks such as malaria detection, prediction of host-pathogen interactions, or drug discovery. Our research aimed to predict the transition of Plasmodium-infected liver cells to the merozoite stage, a key development phase, 15 hours in advance. We collected and analyzed hourly imaging data over a span of at least 38 hours from 400 sequences, encompassing 502 parasites. Our method was compared to human annotations to validate its efficacy. Performance metrics, including the area under the receiver operating characteristic curve (AUC), sensitivity, and specificity, were evaluated on an independent test dataset. The outcomes revealed an AUC of 0.873, a sensitivity of 84.6%, and a specificity of 83.3%, underscoring the potential of our CNN-based framework to predict liver stage development of P. berghei. These findings not only demonstrate the feasibility of our methodology but also could potentially contribute to the broader understanding of parasite biology.
Collapse
Affiliation(s)
- Corin F. Otesteanu
- Artificial Intelligence in Medicine group, University of Bern, Switzerland
| | - Reto Caldelari
- Institute of Cell Biology, University of Bern, Switzerland
| | | | - Raphael Sznitman
- Artificial Intelligence in Medicine group, University of Bern, Switzerland
| |
Collapse
|
6
|
Yanase R, Pruzinova K, Owino BO, Rea E, Moreira-Leite F, Taniguchi A, Nonaka S, Sádlová J, Vojtkova B, Volf P, Sunter JD. Discovery of essential kinetoplastid-insect adhesion proteins and their function in Leishmania-sand fly interactions. Nat Commun 2024; 15:6960. [PMID: 39138209 PMCID: PMC11322530 DOI: 10.1038/s41467-024-51291-z] [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: 09/27/2023] [Accepted: 08/01/2024] [Indexed: 08/15/2024] Open
Abstract
Leishmania species, members of the kinetoplastid parasites, cause leishmaniasis, a neglected tropical disease, in millions of people worldwide. Leishmania has a complex life cycle with multiple developmental forms, as it cycles between a sand fly vector and a mammalian host; understanding their life cycle is critical to understanding disease spread. One of the key life cycle stages is the haptomonad form, which attaches to insect tissues through its flagellum. This adhesion, conserved across kinetoplastid parasites, is implicated in having an important function within their life cycles and hence in disease transmission. Here, we discover the kinetoplastid-insect adhesion proteins (KIAPs), which localise in the attached Leishmania flagellum. Deletion of these KIAPs impairs cell adhesion in vitro and prevents Leishmania from colonising the stomodeal valve in the sand fly, without affecting cell growth. Additionally, loss of parasite adhesion in the sand fly results in reduced physiological changes to the fly, with no observable damage of the stomodeal valve and reduced midgut swelling. These results provide important insights into a comprehensive understanding of the Leishmania life cycle, which will be critical for developing transmission-blocking strategies.
Collapse
Affiliation(s)
- Ryuji Yanase
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK.
- School of Life Sciences, University of Nottingham, Nottingham, UK.
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK.
| | - Katerina Pruzinova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia
| | - Barrack O Owino
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
| | - Edward Rea
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
| | - Flávia Moreira-Leite
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
- Department of Biochemistry, Central Oxford Structural Molecular Imaging Centre (COSMIC), University of Oxford, Oxford, UK
| | - Atsushi Taniguchi
- Laboratory for Spatiotemporal Regulations, National Institute for Basic Biology, Okazaki, Japan
- Research Center of Mathematics for Social Creativity, Research Institute for Electronic Science, Hokkaido University, Sapporo, Japan
| | - Shigenori Nonaka
- Laboratory for Spatiotemporal Regulations, National Institute for Basic Biology, Okazaki, Japan
- Spatiotemporal Regulations Group, Exploratory Research Center for Life and Living Systems, Okazaki, Japan
- Department of Basic Biology, School of Life Science, SOKENDAI, Okazaki, Japan
| | - Jovana Sádlová
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia
| | - Barbora Vojtkova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia
| | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia.
| | - Jack D Sunter
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK.
| |
Collapse
|
7
|
Liu J, Tan YY, Zheng W, Wang Y, Ju LA, Su QP. Nanoscale insights into hematology: super-resolved imaging on blood cell structure, function, and pathology. J Nanobiotechnology 2024; 22:363. [PMID: 38910248 PMCID: PMC11194919 DOI: 10.1186/s12951-024-02605-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/30/2024] [Indexed: 06/25/2024] Open
Abstract
Fluorescence nanoscopy, also known as super-resolution microscopy, has transcended the conventional resolution barriers and enabled visualization of biological samples at nanometric resolutions. A series of super-resolution techniques have been developed and applied to investigate the molecular distribution, organization, and interactions in blood cells, as well as the underlying mechanisms of blood-cell-associated diseases. In this review, we provide an overview of various fluorescence nanoscopy technologies, outlining their current development stage and the challenges they are facing in terms of functionality and practicality. We specifically explore how these innovations have propelled forward the analysis of thrombocytes (platelets), erythrocytes (red blood cells) and leukocytes (white blood cells), shedding light on the nanoscale arrangement of subcellular components and molecular interactions. We spotlight novel biomarkers uncovered by fluorescence nanoscopy for disease diagnosis, such as thrombocytopathies, malignancies, and infectious diseases. Furthermore, we discuss the technological hurdles and chart out prospective avenues for future research directions. This review aims to underscore the significant contributions of fluorescence nanoscopy to the field of blood cell analysis and disease diagnosis, poised to revolutionize our approach to exploring, understanding, and managing disease at the molecular level.
Collapse
Affiliation(s)
- Jinghan Liu
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Yuping Yolanda Tan
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Newtown, NSW, 2042, Australia
| | - Wen Zheng
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Yao Wang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Newtown, NSW, 2042, Australia
| | - Qian Peter Su
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, 2007, Australia.
- Heart Research Institute, Newtown, NSW, 2042, Australia.
| |
Collapse
|
8
|
Thiele PJ, Mela-Lopez R, Blandin SA, Klug D. Let it glow: genetically encoded fluorescent reporters in Plasmodium. Malar J 2024; 23:114. [PMID: 38643106 PMCID: PMC11032601 DOI: 10.1186/s12936-024-04936-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/06/2024] [Indexed: 04/22/2024] Open
Abstract
The use of fluorescent proteins (FPs) in Plasmodium parasites has been key to understand the biology of this obligate intracellular protozoon. FPs like the green fluorescent protein (GFP) enabled to explore protein localization, promoter activity as well as dynamic processes like protein export and endocytosis. Furthermore, FP biosensors have provided detailed information on physiological parameters at the subcellular level, and fluorescent reporter lines greatly extended the malariology toolbox. Still, in order to achieve optimal results, it is crucial to know exactly the properties of the FP of choice and the genetic scenario in which it will be used. This review highlights advantages and disadvantages of available landing sites and promoters that have been successfully applied for the ectopic expression of FPs in Plasmodium berghei and Plasmodium falciparum. Furthermore, the properties of newly developed FPs beyond DsRed and EGFP, in the visualization of cells and cellular structures as well as in the sensing of small molecules are discussed.
Collapse
Affiliation(s)
- Pia J Thiele
- Inserm, CNRS, Université de Strasbourg, UPR9022/U1257, Mosquito Immune Responses (MIR), IBMC, F-67000, Strasbourg, France
| | - Raquel Mela-Lopez
- Inserm, CNRS, Université de Strasbourg, UPR9022/U1257, Mosquito Immune Responses (MIR), IBMC, F-67000, Strasbourg, France
| | - Stéphanie A Blandin
- Inserm, CNRS, Université de Strasbourg, UPR9022/U1257, Mosquito Immune Responses (MIR), IBMC, F-67000, Strasbourg, France
| | - Dennis Klug
- Inserm, CNRS, Université de Strasbourg, UPR9022/U1257, Mosquito Immune Responses (MIR), IBMC, F-67000, Strasbourg, France.
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps University Marburg, 35037, Marburg, Germany.
| |
Collapse
|
9
|
Yeh YT, Del Álamo JC, Caffrey CR. Biomechanics of parasite migration within hosts. Trends Parasitol 2024; 40:164-175. [PMID: 38172015 DOI: 10.1016/j.pt.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024]
Abstract
The dissemination of protozoan and metazoan parasites through host tissues is hindered by cellular barriers, dense extracellular matrices, and fluid forces in the bloodstream. To overcome these diverse biophysical impediments, parasites implement versatile migratory strategies. Parasite-exerted mechanical forces and upregulation of the host's cellular contractile machinery are the motors for these strategies, and these are comparably better characterized for protozoa than for helminths. Using the examples of the protozoans, Toxoplasma gondii and Plasmodium, and the metazoan, Schistosoma mansoni, we highlight how quantitative tools such as traction force and reflection interference contrast microscopies have improved our understanding of how parasites alter host mechanobiology to promote their migration.
Collapse
Affiliation(s)
- Yi-Ting Yeh
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98109, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA.
| | - Juan C Del Álamo
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98109, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA; Division of Cardiology, University of Washington, Seattle, WA 98109, USA; Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA 93093, USA
| | - Conor R Caffrey
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, 9500 Gilman Drive, MC0657, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
10
|
Pietsch E, Ramaprasad A, Bielfeld S, Wohlfarter Y, Maco B, Niedermüller K, Wilcke L, Kloehn J, Keller MA, Soldati-Favre D, Blackman MJ, Gilberger TW, Burda PC. A patatin-like phospholipase is important for mitochondrial function in malaria parasites. mBio 2023; 14:e0171823. [PMID: 37882543 PMCID: PMC10746288 DOI: 10.1128/mbio.01718-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/12/2023] [Indexed: 10/27/2023] Open
Abstract
IMPORTANCE For their proliferation within red blood cells, malaria parasites depend on a functional electron transport chain (ETC) within their mitochondrion, which is the target of several antimalarial drugs. Here, we have used gene disruption to identify a patatin-like phospholipase, PfPNPLA2, as important for parasite replication and mitochondrial function in Plasmodium falciparum. Parasites lacking PfPNPLA2 show defects in their ETC and become hypersensitive to mitochondrion-targeting drugs. Furthermore, PfPNPLA2-deficient parasites show differences in the composition of their cardiolipins, a unique class of phospholipids with key roles in mitochondrial functions. Finally, we demonstrate that parasites devoid of PfPNPLA2 have a defect in gametocyte maturation, underlining the importance of a functional ETC for parasite transmission to the mosquito vector.
Collapse
Affiliation(s)
- Emma Pietsch
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- University of Hamburg, Hamburg, Germany
| | - Abhinay Ramaprasad
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Sabrina Bielfeld
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- University of Hamburg, Hamburg, Germany
| | - Yvonne Wohlfarter
- Institute of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Bohumil Maco
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Korbinian Niedermüller
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- University of Hamburg, Hamburg, Germany
| | - Louisa Wilcke
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- University of Hamburg, Hamburg, Germany
| | - Joachim Kloehn
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Markus A. Keller
- Institute of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Michael J. Blackman
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Tim-Wolf Gilberger
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- University of Hamburg, Hamburg, Germany
| | - Paul-Christian Burda
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- University of Hamburg, Hamburg, Germany
| |
Collapse
|
11
|
Stürmer VS, Stopper S, Binder P, Klemmer A, Lichti NP, Becker NB, Guizetti J. Progeny counter mechanism in malaria parasites is linked to extracellular resources. PLoS Pathog 2023; 19:e1011807. [PMID: 38051755 PMCID: PMC10723702 DOI: 10.1371/journal.ppat.1011807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 12/15/2023] [Accepted: 11/07/2023] [Indexed: 12/07/2023] Open
Abstract
Malaria is caused by the rapid proliferation of Plasmodium parasites in patients and disease severity correlates with the number of infected red blood cells in circulation. Parasite multiplication within red blood cells is called schizogony and occurs through an atypical multinucleated cell division mode. The mechanisms regulating the number of daughter cells produced by a single progenitor are poorly understood. We investigated underlying regulatory principles by quantifying nuclear multiplication dynamics in Plasmodium falciparum and knowlesi using super-resolution time-lapse microscopy. This confirmed that the number of daughter cells was consistent with a model in which a counter mechanism regulates multiplication yet incompatible with a timer mechanism. P. falciparum cell volume at the start of nuclear division correlated with the final number of daughter cells. As schizogony progressed, the nucleocytoplasmic volume ratio, which has been found to be constant in all eukaryotes characterized so far, increased significantly, possibly to accommodate the exponentially multiplying nuclei. Depleting nutrients by dilution of culture medium caused parasites to produce fewer merozoites and reduced proliferation but did not affect cell volume or total nuclear volume at the end of schizogony. Our findings suggest that the counter mechanism implicated in malaria parasite proliferation integrates extracellular resource status to modify progeny number during blood stage infection.
Collapse
Affiliation(s)
- Vanessa S. Stürmer
- Heidelberg University Medical Faculty, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Sophie Stopper
- Heidelberg University Medical Faculty, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Patrick Binder
- Institute for Theoretical Physics and BioQuant, Heidelberg University, Heidelberg, Germany
- Theoretical Systems Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anja Klemmer
- Heidelberg University Medical Faculty, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Nicolas P. Lichti
- Heidelberg University Medical Faculty, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Nils B. Becker
- Theoretical Systems Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julien Guizetti
- Heidelberg University Medical Faculty, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
12
|
Vigetti L, Tardieux I. Fostering innovation to solve the biomechanics of microbe-host interactions: Focus on the adhesive forces underlying Apicomplexa parasite biology. Biol Cell 2023; 115:e202300016. [PMID: 37227253 DOI: 10.1111/boc.202300016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 05/26/2023]
Abstract
The protozoa, Toxoplasma gondii and Plasmodium spp., are preeminent members of the Apicomplexa parasitic phylum in large part due to their public health and economic impact. Hence, they serve as model unicellular eukaryotes with which to explore the repertoire of molecular and cellular strategies that specific developmental morphotypes deploy to timely adjust to their host(s) in order to perpetuate. In particular, host tissue- and cell-invasive morphotypes termed zoites alternate extracellular and intracellular lifestyles, thereby sensing and reacting to a wealth of host-derived biomechanical cues over their partnership. In the recent years, biophysical tools especially related to real time force measurement have been introduced, teaching us how creative are these microbes to shape a unique motility system that powers fast gliding through a variety of extracellular matrices, across cellular barriers, in vascular systems or into host cells. Equally performant was this toolkit to start illuminating how parasites manipulate their hosting cell adhesive and rheological properties to their advantage. In this review, besides highlighting major discoveries along the way, we discuss the most promising development, synergy, and multimodal integration in active noninvasive force microscopy methods. These should in the near future unlock current limitations and allow capturing, from molecules to tissues, the many biomechanical and biophysical interplays over the dynamic host and microbe partnership.
Collapse
Affiliation(s)
- Luis Vigetti
- Team Biomechanics of Host-Parasite Interactions, Institute for Advanced Biosciences, CNRS UMR5309, INSERM U1209, University of Grenoble Alpes, Grenoble, France
| | - Isabelle Tardieux
- Team Biomechanics of Host-Parasite Interactions, Institute for Advanced Biosciences, CNRS UMR5309, INSERM U1209, University of Grenoble Alpes, Grenoble, France
| |
Collapse
|
13
|
Burda PC, Ramaprasad A, Bielfeld S, Pietsch E, Woitalla A, Söhnchen C, Singh MN, Strauss J, Sait A, Collinson LM, Schwudke D, Blackman MJ, Gilberger TW. Global analysis of putative phospholipases in Plasmodium falciparum reveals an essential role of the phosphoinositide-specific phospholipase C in parasite maturation. mBio 2023; 14:e0141323. [PMID: 37489900 PMCID: PMC10470789 DOI: 10.1128/mbio.01413-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 07/26/2023] Open
Abstract
For its replication within red blood cells, the malaria parasite depends on a highly active and regulated lipid metabolism. Enzymes involved in lipid metabolic processes such as phospholipases are, therefore, potential drug targets. Here, using reverse genetics approaches, we show that only 1 out of the 19 putative phospholipases expressed in asexual blood stages of Plasmodium falciparum is essential for proliferation in vitro, pointing toward a high level of redundancy among members of this enzyme family. Using conditional mislocalization and gene disruption techniques, we show that this essential phosphoinositide-specific phospholipase C (PI-PLC, PF3D7_1013500) has a previously unrecognized essential role during intracellular parasite maturation, long before its previously perceived role in parasite egress and invasion. Subsequent lipidomic analysis suggests that PI-PLC mediates cleavage of phosphatidylinositol bisphosphate (PIP2) in schizont-stage parasites, underlining its critical role in regulating phosphoinositide levels in the parasite. IMPORTANCE The clinical symptoms of malaria arise due to repeated rounds of replication of Plasmodium parasites within red blood cells (RBCs). Central to this is an intense period of membrane biogenesis. Generation of membranes not only requires de novo synthesis and acquisition but also the degradation of phospholipids, a function that is performed by phospholipases. In this study, we investigate the essentiality of the 19 putative phospholipase enzymes that the human malaria parasite Plasmodium falciparum expresses during its replication within RBCs. We not only show that a high level of functional redundancy exists among these enzymes but, at the same time, also identify an essential role for the phosphoinositide-specific phospholipase C in parasite development and cleavage of the phospholipid phosphatidylinositol bisphosphate.
Collapse
Affiliation(s)
- Paul-Christian Burda
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- University of Hamburg, Hamburg, Germany
| | - Abhinay Ramaprasad
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Sabrina Bielfeld
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- University of Hamburg, Hamburg, Germany
| | - Emma Pietsch
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- University of Hamburg, Hamburg, Germany
| | - Anna Woitalla
- Division of Bioanalytical Chemistry, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Christoph Söhnchen
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- University of Hamburg, Hamburg, Germany
| | - Mehar Nihal Singh
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Jan Strauss
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- University of Hamburg, Hamburg, Germany
| | - Aaron Sait
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Lucy M. Collinson
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Dominik Schwudke
- Division of Bioanalytical Chemistry, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
- German Center for Infection Research, Thematic Translational Unit Tuberculosis, Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
- German Center for Lung Research (DZL), Airway Research Center North (ARCN), Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Michael J. Blackman
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Tim-Wolf Gilberger
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- University of Hamburg, Hamburg, Germany
| |
Collapse
|
14
|
Cooper C, Thompson RCA, Clode PL. Investigating parasites in three dimensions: trends in volume microscopy. Trends Parasitol 2023; 39:668-681. [PMID: 37302958 DOI: 10.1016/j.pt.2023.05.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/17/2023] [Accepted: 05/17/2023] [Indexed: 06/13/2023]
Abstract
To best understand parasite, host, and vector morphologies, host-parasite interactions, and to develop new drug and vaccine targets, structural data should, ideally, be obtained and visualised in three dimensions (3D). Recently, there has been a significant uptake of available 3D volume microscopy techniques that allow collection of data across centimetre (cm) to Angstrom (Å) scales by utilising light, X-ray, electron, and ion sources. Here, we present and discuss microscopy tools available for the collection of 3D structural data, focussing on electron microscopy-based techniques. We highlight their strengths and limitations, such that parasitologists can identify techniques best suited to answer their research questions. Additionally, we review the importance of volume microscopy to the advancement of the field of parasitology.
Collapse
Affiliation(s)
- Crystal Cooper
- Centre for Microscopy, Characterisation, and Analysis, University of Western Australia, Stirling Hwy, Crawley, WA 6009, Australia.
| | - R C Andrew Thompson
- School of Veterinary and Life Sciences, Murdoch University, 90 South Street, Murdoch, WA 6150, Australia
| | - Peta L Clode
- Centre for Microscopy, Characterisation, and Analysis, University of Western Australia, Stirling Hwy, Crawley, WA 6009, Australia; School of Biological Sciences, University of Western Australia, Stirling Hwy, Crawley, WA 6009, Australia
| |
Collapse
|
15
|
Kehrer J, Pietsch E, Heinze J, Spielmann T, Frischknecht F. Clearing of hemozoin crystals in malaria parasites enables whole-cell STED microscopy. J Cell Sci 2023; 136:286288. [PMID: 36511329 DOI: 10.1242/jcs.260399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022] Open
Abstract
Malaria is a devastating mosquito-borne parasitic disease that manifests when Plasmodium parasites replicate within red blood cells. During the development within the red blood cell, the parasite digests hemoglobin and crystalizes the otherwise toxic heme. The resulting hemozoin crystals limit imaging by STED nanoscopy owing to their high light-absorbing capacity, which leads to immediate cell destruction upon contact with the laser. Here, we establish CUBIC-P-based clearing of hemozoin crystals, enabling whole-cell STED nanoscopy of parasites within red blood cells. Hemozoin-cleared infected red blood cells could reliably be stained with antibodies, and hence proteins in the hemozoin-containing digestive vacuole membrane, as well as in secretory vesicles of gametocytes, could be imaged at high resolution. Thus, this process is a valuable tool to study and understand parasite biology and the potential molecular mechanisms mediating drug resistance. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Jessica Kehrer
- Integrative Parasitology, Center for Integrative Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 344, 69120 Heidelberg, Germany.,German Center for Infection Research, DZIF, partner site Heidelberg, 69120 Heidelberg, Germany.,Infectious Diseases Imaging Platform, Center for Integrative Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 344, 69120 Heidelberg, Germany
| | - Emma Pietsch
- Integrative Parasitology, Center for Integrative Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 344, 69120 Heidelberg, Germany
| | - Julia Heinze
- Integrative Parasitology, Center for Integrative Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 344, 69120 Heidelberg, Germany
| | - Tobias Spielmann
- Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht Str. 74, 20359 Hamburg, Germany
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Integrative Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 344, 69120 Heidelberg, Germany.,German Center for Infection Research, DZIF, partner site Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
16
|
Shao J. Labeling Strategies for Surface-Exposed Protein Visualization and Determination in Plasmodium falciparum Malaria. Front Cell Infect Microbiol 2022; 12:914297. [PMID: 35755836 PMCID: PMC9226428 DOI: 10.3389/fcimb.2022.914297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jinfeng Shao
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| |
Collapse
|
17
|
Kent RS, Briggs EM, Colon BL, Alvarez C, Silva Pereira S, De Niz M. Paving the Way: Contributions of Big Data to Apicomplexan and Kinetoplastid Research. Front Cell Infect Microbiol 2022; 12:900878. [PMID: 35734575 PMCID: PMC9207352 DOI: 10.3389/fcimb.2022.900878] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
In the age of big data an important question is how to ensure we make the most out of the resources we generate. In this review, we discuss the major methods used in Apicomplexan and Kinetoplastid research to produce big datasets and advance our understanding of Plasmodium, Toxoplasma, Cryptosporidium, Trypanosoma and Leishmania biology. We debate the benefits and limitations of the current technologies, and propose future advancements that may be key to improving our use of these techniques. Finally, we consider the difficulties the field faces when trying to make the most of the abundance of data that has already been, and will continue to be, generated.
Collapse
Affiliation(s)
- Robyn S. Kent
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT, United States
| | - Emma M. Briggs
- Institute for Immunology and Infection Research, School of Biological Sciences, University Edinburgh, Edinburgh, United Kingdom
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Beatrice L. Colon
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Catalina Alvarez
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Sara Silva Pereira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Mariana De Niz
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
- Institut Pasteur, Paris, France
| |
Collapse
|
18
|
Abstract
Blood cell analysis is essential for the diagnosis and identification of hematological malignancies. The use of digital microscopy systems has been extended in clinical laboratories. Super-resolution microscopy (SRM) has attracted wide attention in the medical field due to its nanoscale spatial resolution and high sensitivity. It is considered to be a potential method of blood cell analysis that may have more advantages than traditional approaches such as conventional optical microscopy and hematology analyzers in certain examination projects. In this review, we firstly summarize several common blood cell analysis technologies in the clinic, and analyze the advantages and disadvantages of these technologies. Then, we focus on the basic principles and characteristics of three representative SRM techniques, as well as the latest advances in these techniques for blood cell analysis. Finally, we discuss the developmental trend and possible research directions of SRM, and provide some discussions on further development of technologies for blood cell analysis.
Collapse
|
19
|
Edgar RCS, Counihan NA, McGowan S, de Koning-Ward TF. Methods Used to Investigate the Plasmodium falciparum Digestive Vacuole. Front Cell Infect Microbiol 2022; 11:829823. [PMID: 35096663 PMCID: PMC8794586 DOI: 10.3389/fcimb.2021.829823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 12/22/2021] [Indexed: 11/13/2022] Open
Abstract
Plasmodium falciparum malaria remains a global health problem as parasites continue to develop resistance to all antimalarials in use. Infection causes clinical symptoms during the intra-erythrocytic stage of the lifecycle where the parasite infects and replicates within red blood cells (RBC). During this stage, P. falciparum digests the main constituent of the RBC, hemoglobin, in a specialized acidic compartment termed the digestive vacuole (DV), a process essential for survival. Many therapeutics in use target one or multiple aspects of the DV, with chloroquine and its derivatives, as well as artemisinin, having mechanisms of action within this organelle. In order to better understand how current therapeutics and those under development target DV processes, techniques used to investigate the DV are paramount. This review outlines the involvement of the DV in therapeutics currently in use and focuses on the range of techniques that are currently utilized to study this organelle including microscopy, biochemical analysis, genetic approaches and metabolomic studies. Importantly, continued development and application of these techniques will aid in our understanding of the DV and in the development of new therapeutics or therapeutic partners for the future.
Collapse
Affiliation(s)
- Rebecca C. S. Edgar
- School of Medicine, Deakin University, Geelong, VIC, Australia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| | - Natalie A. Counihan
- School of Medicine, Deakin University, Geelong, VIC, Australia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| | - Sheena McGowan
- Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, VIC, Australia
- Centre to Impact AMR, Monash University, Monash University, Clayton, VIC, Australia
| | - Tania F. de Koning-Ward
- School of Medicine, Deakin University, Geelong, VIC, Australia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
20
|
Kaur M, Yardley V, Wang K, Masania J, Arroo RRJ, Turner DB, Li M. Artemisinin Cocrystals for Bioavailability Enhancement. Part 2: In Vivo Bioavailability and Physiologically Based Pharmacokinetic Modeling. Mol Pharm 2021; 18:4272-4289. [PMID: 34748332 DOI: 10.1021/acs.molpharmaceut.1c00385] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We report the evaluation and prediction of the pharmacokinetic (PK) performance of artemisinin (ART) cocrystal formulations, that is, 1:1 artemisinin/orcinol (ART-ORC) and 2:1 artemisinin/resorcinol (ART2-RES), using in vivo murine animal and physiologically based pharmacokinetic (PBPK) models. The efficacy of the ART cocrystal formulations along with the parent drug ART was tested in mice infected with Plasmodium berghei. When given at the same dose, the ART cocrystal formulation showed a significant reduction in parasitaemia at day 4 after infection compared to ART alone. PK parameters including Cmax (maximum plasma concentration), Tmax (time to Cmax), and AUC (area under the curve) were obtained by determining drug concentrations in the plasma using liquid chromatography-high-resolution mass spectrometry (LC-HRMS), showing enhanced ART levels after dosage with the cocrystal formulations. The dose-response tests revealed that a significantly lower dose of the ART cocrystals in the formulation was required to achieve a similar therapeutic effect as ART alone. A PBPK model was developed using a PBPK mouse simulator to accurately predict the in vivo behavior of the cocrystal formulations by combining in vitro dissolution profiles with the properties of the parent drug ART. The study illustrated that information from classical in vitro and in vivo experimental investigations of the parent drug of ART formulations can be coupled with PBPK modeling to predict the PK parameters of an ART cocrystal formulation in an efficient manner. Therefore, the proposed modeling strategy could be used to establish in vitro and in vivo correlations for different cocrystals intended to improve dissolution properties and to support clinical candidate selection, contributing to the assessment of cocrystal developability and formulation development.
Collapse
Affiliation(s)
- Manreet Kaur
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, U.K
| | - Vanessa Yardley
- Department of Infection & Immunity, Faculty of Infectious & Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, U.K
| | - Ke Wang
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, U.K
| | - Jinit Masania
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, U.K
| | - Randolph R J Arroo
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, U.K
| | - David B Turner
- Certara UK Limited, Simcyp Division, Sheffield S1 2BJ, U.K
| | - Mingzhong Li
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, U.K
| |
Collapse
|
21
|
Ressurreição M, van Ooij C. Lipid transport proteins in malaria, from Plasmodium parasites to their hosts. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:159047. [PMID: 34461309 DOI: 10.1016/j.bbalip.2021.159047] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 08/08/2021] [Accepted: 08/10/2021] [Indexed: 11/25/2022]
Abstract
Eukaryotic unicellular pathogens from the genus Plasmodium are the etiological agents of malaria, a disease that persists over a wide range of vertebrate species, including humans. During its dynamic lifecycle, survival in the different hosts depends on the parasite's ability to establish a suitable environmental milieu. To achieve this, specific host processes are exploited to support optimal growth, including extensive modifications to the infected host cell. These modifications include the formation of novel membranous structures, which are induced by the parasite. Consequently, to maintain a finely tuned and dynamic lipid environment, the organisation and distribution of lipids to different cell sites likely requires specialised lipid transfer proteins (LTPs). Indeed, several parasite and host-derived LTPs have been identified and shown to be essential at specific stages. Here we describe the roles of LTPs in parasite development and adaptation to its host including how the latest studies are profiting from the improved genetic, lipidomic and imaging toolkits available to study Plasmodium parasites. Lastly, a list of predicted Plasmodium LTPs is provided to encourage research in this field.
Collapse
Affiliation(s)
- Margarida Ressurreição
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom.
| | - Christiaan van Ooij
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom.
| |
Collapse
|
22
|
Simon CS, Stürmer VS, Guizetti J. How Many Is Enough? - Challenges of Multinucleated Cell Division in Malaria Parasites. Front Cell Infect Microbiol 2021; 11:658616. [PMID: 34026661 PMCID: PMC8137892 DOI: 10.3389/fcimb.2021.658616] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/21/2021] [Indexed: 12/12/2022] Open
Abstract
Regulating the number of progeny generated by replicative cell cycles is critical for any organism to best adapt to its environment. Classically, the decision whether to divide further is made after cell division is completed by cytokinesis and can be triggered by intrinsic or extrinsic factors. Contrarily, cell cycles of some species, such as the malaria-causing parasites, go through multinucleated cell stages. Hence, their number of progeny is determined prior to the completion of cell division. This should fundamentally affect how the process is regulated and raises questions about advantages and challenges of multinucleation in eukaryotes. Throughout their life cycle Plasmodium spp. parasites undergo four phases of extensive proliferation, which differ over three orders of magnitude in the amount of daughter cells that are produced by a single progenitor. Even during the asexual blood stage proliferation parasites can produce very variable numbers of progeny within one replicative cycle. Here, we review the few factors that have been shown to affect those numbers. We further provide a comparative quantification of merozoite numbers in several P. knowlesi and P. falciparum parasite strains, and we discuss the general processes that may regulate progeny number in the context of host-parasite interactions. Finally, we provide a perspective of the critical knowledge gaps hindering our understanding of the molecular mechanisms underlying this exciting and atypical mode of parasite multiplication.
Collapse
Affiliation(s)
| | | | - Julien Guizetti
- Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
23
|
Bertiaux E, Balestra AC, Bournonville L, Louvel V, Maco B, Soldati-Favre D, Brochet M, Guichard P, Hamel V. Expansion microscopy provides new insights into the cytoskeleton of malaria parasites including the conservation of a conoid. PLoS Biol 2021; 19:e3001020. [PMID: 33705377 PMCID: PMC7951857 DOI: 10.1371/journal.pbio.3001020] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 01/21/2021] [Indexed: 12/17/2022] Open
Abstract
Malaria is caused by unicellular Plasmodium parasites. Plasmodium relies on diverse microtubule cytoskeletal structures for its reproduction, multiplication, and dissemination. Due to the small size of this parasite, its cytoskeleton has been primarily observable by electron microscopy (EM). Here, we demonstrate that the nanoscale cytoskeleton organisation is within reach using ultrastructure expansion microscopy (U-ExM). In developing microgametocytes, U-ExM allows monitoring the dynamic assembly of axonemes and concomitant tubulin polyglutamylation in whole cells. In the invasive merozoite and ookinete forms, U-ExM unveils the diversity across Plasmodium stages and species of the subpellicular microtubule arrays that confer cell rigidity. In ookinetes, we additionally identify an apical tubulin ring (ATR) that colocalises with markers of the conoid in related apicomplexan parasites. This tubulin-containing structure was presumed to be lost in Plasmodium despite its crucial role in motility and invasion in other apicomplexans. Here, U-ExM reveals that a divergent and considerably reduced form of the conoid is actually conserved in Plasmodium species.
Collapse
Affiliation(s)
- Eloïse Bertiaux
- University of Geneva, Department of Cell Biology, Faculty of Science, Geneva, Switzerland
| | - Aurélia C. Balestra
- University of Geneva, Department of Microbiology and Molecular Medicine, Faculty of Medicine, Geneva, Switzerland
| | - Lorène Bournonville
- University of Geneva, Department of Cell Biology, Faculty of Science, Geneva, Switzerland
| | - Vincent Louvel
- University of Geneva, Department of Cell Biology, Faculty of Science, Geneva, Switzerland
| | - Bohumil Maco
- University of Geneva, Department of Microbiology and Molecular Medicine, Faculty of Medicine, Geneva, Switzerland
| | - Dominique Soldati-Favre
- University of Geneva, Department of Microbiology and Molecular Medicine, Faculty of Medicine, Geneva, Switzerland
| | - Mathieu Brochet
- University of Geneva, Department of Microbiology and Molecular Medicine, Faculty of Medicine, Geneva, Switzerland
| | - Paul Guichard
- University of Geneva, Department of Cell Biology, Faculty of Science, Geneva, Switzerland
| | - Virginie Hamel
- University of Geneva, Department of Cell Biology, Faculty of Science, Geneva, Switzerland
| |
Collapse
|
24
|
Lu L, Sun Y, Wan C, Hu Y, Lo PC, Lovell JF, Yang K, Jin H. Role of intravital imaging in nanomedicine-assisted anti-cancer therapy. Curr Opin Biotechnol 2021; 69:153-161. [PMID: 33476937 DOI: 10.1016/j.copbio.2020.12.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 12/22/2020] [Accepted: 12/27/2020] [Indexed: 01/07/2023]
Abstract
Although nanomedicines have provided promising anti-tumor effects in cancer animal models, their clinical success remains limited. One of the most significant barriers in the clinical translation of nanomedicines is that they consist of multiple components, each of which may have different toxicities and therapeutic effects. Intravital imaging provides high spatial and temporal resolution for visualizing nanomedicine-mediated interactions between immune cells and tumor cells in real-time. Intravital imaging can facilitate the in vivo evaluation of the properties and effects of nanomedicines, such as their ability to cross the tumor vasculature, specifically eliminate the cancer cells, and modulate the immune cells found in the tumor microenvironment (TME). Thus, intravital imaging can provide direct evidence of nanomedicine's intravital behavior to better understand mechanism and accelerate clinical translation. In this review, we summarize several applications and latest advances in intravital imaging in nanomedicine-assisted anti-cancer therapy and discuss future perspectives in the field.
Collapse
Affiliation(s)
- Lisen Lu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yajie Sun
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chao Wan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yan Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Pui-Chi Lo
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Kunyu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Honglin Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
25
|
|
26
|
Guo W, Kanski M, Liu W, Gołuński M, Zhou Y, Wang Y, Cheng C, Du Y, Postawa Z, Wei WD, Zhu Z. Three-Dimensional Mass Spectrometric Imaging of Biological Structures Using a Vacuum-Compatible Microfluidic Device. Anal Chem 2020; 92:13785-13793. [PMID: 32872776 DOI: 10.1021/acs.analchem.0c02204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Three-dimensional (3D) molecular imaging of biological structures is important for a wide range of research. In recent decades, secondary-ion mass spectrometry (SIMS) has been recognized as a powerful technique for both two-dimensional and 3D molecular imaging. Sample fixations (e.g., chemical fixation and cryogenic fixation methods) are necessary to adapt biological samples to the vacuum condition in the SIMS chamber, which has been demonstrated to be nontrivial and less controllable, thus limiting the wider application of SIMS on 3D molecular analysis of biological samples. Our group recently developed in situ liquid SIMS that offers great opportunities for the molecular study of various liquids and liquid interfaces. In this work, we demonstrate that a further development of the vacuum-compatible microfluidic device used in in situ liquid SIMS provides a convenient freeze-fixation of biological samples and leads to more controllable and convenient 3D molecular imaging. The special design of this new vacuum-compatible liquid chamber allows an easy determination of sputter rates of ice, which is critical for calibrating the depth scale of frozen biological samples. Sputter yield of a 20 keV Ar1800+ ion on ice has been determined as 1500 (±8%) water molecules per Ar1800+ ion, consistent with our results from molecular dynamics simulations. Moreover, using the information of ice sputter yield, we successfully conduct 3D molecular imaging of frozen homogenized milk and observe network structures of interesting organic and inorganic species. Taken together, our results will significantly benefit various research fields relying on 3D molecular imaging of biological structures.
Collapse
Affiliation(s)
- Wenxiao Guo
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99354, United States.,Department of Chemistry and Center for Catalysis, University of Florida, Gainesville, Florida 32611, United States
| | - Michal Kanski
- Smoluchowski Institute of Physics, Jagiellonian University, S. Lojasiewicza 11, Kraków 31-007, Poland
| | - Wen Liu
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99354, United States
| | - Mikołaj Gołuński
- Smoluchowski Institute of Physics, Jagiellonian University, S. Lojasiewicza 11, Kraków 31-007, Poland
| | - Yadong Zhou
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99354, United States
| | - Yining Wang
- Physical and Computational Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99354, United States
| | - Cuixia Cheng
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99354, United States
| | - Yingge Du
- Physical and Computational Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99354, United States
| | - Zbigniew Postawa
- Smoluchowski Institute of Physics, Jagiellonian University, S. Lojasiewicza 11, Kraków 31-007, Poland
| | - Wei David Wei
- Department of Chemistry and Center for Catalysis, University of Florida, Gainesville, Florida 32611, United States
| | - Zihua Zhu
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99354, United States
| |
Collapse
|
27
|
Wilairat P, Auparakkitanon S. Plug for the parasitophorous duct: a solution of two conundra. Malar J 2020; 19:370. [PMID: 33066767 PMCID: PMC7566142 DOI: 10.1186/s12936-020-03445-9] [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: 08/24/2020] [Accepted: 10/10/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We present two conundra in the biology of intraerythrocytic malaria parasite: how an apparent open parasitophorous duct provide direct access of only a select set of serum proteins to the parasitophorous vacuole, and how proteases mediate membrane lysis to allow merozoite egress. SOLUTION We posit the existence of a parasitophorous vacuolar duct plug that is originally formed from a tight junction (or parts thereof) between merozoite apical surface and red blood cell plasma membrane, which by moving over the parasite surface towards the posterior end draws the parasite into the host cell interior, and by remaining at the passage orifice provides a location of transporter(s) for import of serum proteins into parasitophorous vacuole and an opening for merozoite egress upon its dissolution/dismantling through protease(s) action. CONCLUSION This notion obviates the need of a distinct intact parasitophorous vacuolar membrane, which in the proposed model is an extension of the red blood cell membrane but still forms an intracellular compartment for parasite growth and development. The model is testable using existing high-resolution electron and X-ray tomography tools.
Collapse
Affiliation(s)
- Prapon Wilairat
- Department of Biochemistry, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok, 10400, Thailand.
| | - Saranya Auparakkitanon
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama 6 Road, Bangkok, 10400, Thailand
| |
Collapse
|
28
|
González LM, Estrada K, Grande R, Jiménez-Jacinto V, Vega-Alvarado L, Sevilla E, de la Barrera J, Cuesta I, Zaballos Á, Bautista JM, Lobo CA, Sánchez-Flores A, Montero E. Comparative and functional genomics of the protozoan parasite Babesia divergens highlighting the invasion and egress processes. PLoS Negl Trop Dis 2019; 13:e0007680. [PMID: 31425518 PMCID: PMC6715253 DOI: 10.1371/journal.pntd.0007680] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 08/29/2019] [Accepted: 08/01/2019] [Indexed: 12/31/2022] Open
Abstract
Babesiosis is considered an emerging disease because its incidence has significantly increased in the last 30 years, providing evidence of the expanding range of this rare but potentially life-threatening zoonotic disease. Babesia divergens is a causative agent of babesiosis in humans and cattle in Europe. The recently sequenced genome of B. divergens revealed over 3,741 protein coding-genes and the 10.7-Mb high-quality draft become the first reference tool to study the genome structure of B. divergens. Now, by exploiting this sequence data and using new computational tools and assembly strategies, we have significantly improved the quality of the B. divergens genome. The new assembly shows better continuity and has a higher correspondence to B. bovis chromosomes. Moreover, we present a differential expression analysis using RNA sequencing of the two different stages of the asexual lifecycle of B. divergens: the free merozoite capable of invading erythrocytes and the intraerythrocytic parasite stage that remains within the erythrocyte until egress. Comparison of mRNA levels of both stages identified 1,441 differentially expressed genes. From these, around half were upregulated and the other half downregulated in the intraerythrocytic stage. Orthogonal validation by real-time quantitative reverse transcription PCR confirmed the differential expression. A moderately increased expression level of genes, putatively involved in the invasion and egress processes, were revealed in the intraerythrocytic stage compared with the free merozoite. On the basis of these results and in the absence of molecular models of invasion and egress for B. divergens, we have proposed the identified genes as putative molecular players in the invasion and egress processes. Our results contribute to an understanding of key parasitic strategies and pathogenesis and could be a valuable genomic resource to exploit for the design of diagnostic methods, drugs and vaccines to improve the control of babesiosis. Babesiosis has long been recognized as an economically important disease of cattle, but only in the last 40 years has Babesia been recognized as an important pathogen in humans. Babesiosis in humans is caused by one of several species (B. microti, B. divergens, B. duncani and B. venatorum). The complete Babesia lifecycle requires two hosts, the ixodid ticks and a vertebrate host. It is the parasite's ability to first recognize and then invade host erythrocytes that is central to the pathogenesis of babesiosis. Once inside the cell, the parasite begins a cycle of maturation and growth, resulting in merozoites that egress from the red blood cells (RBCs) and seek new, uninfected RBCs to invade, perpetuating the infection. To better understand this asexual lifecycle, the authors focused on the parasite genome and transcriptome of the asexual erythrocytic forms of B. divergens. Through this functional and comparative genomic approach, the authors have identified genes putatively involved in invasion, gliding motility, moving junction formation and egress, providing new insights into the molecular mechanisms of these processes necessary for B. divergens to survive and propagate during its life cycle.
Collapse
Affiliation(s)
- Luis Miguel González
- Laboratorio de Referencia e Investigación en Parasitología, Centro Nacional de Microbiología, ISCIII Majadahonda, Madrid, Spain
| | - Karel Estrada
- Unidad Universitaria de Secuenciación Masiva y Bioinformática, Instituto de Biotecnología, Cuernavaca, México
| | - Ricardo Grande
- Unidad Universitaria de Secuenciación Masiva y Bioinformática, Instituto de Biotecnología, Cuernavaca, México
| | - Verónica Jiménez-Jacinto
- Unidad Universitaria de Secuenciación Masiva y Bioinformática, Instituto de Biotecnología, Cuernavaca, México
| | | | - Elena Sevilla
- Laboratorio de Referencia e Investigación en Parasitología, Centro Nacional de Microbiología, ISCIII Majadahonda, Madrid, Spain
| | - Jorge de la Barrera
- Unidad de Bioinformática, Área de Unidades Centrales Científico-Técnicas, ISCIII, Majadahonda, Madrid, Spain
| | - Isabel Cuesta
- Unidad de Bioinformática, Área de Unidades Centrales Científico-Técnicas, ISCIII, Majadahonda, Madrid, Spain
| | - Ángel Zaballos
- Unidad de Genómica, Área de Unidades Centrales Científico-Técnicas, ISCIII, Majadahonda, Madrid, Spain
| | - José Manuel Bautista
- Department of Biochemistry and Molecular Biology & Research Institute Hospital 12 de Octubre, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Cheryl A. Lobo
- Blood Borne Parasites, LFKRI, New York Blood Center, New York, New York, United States of America
| | - Alejandro Sánchez-Flores
- Unidad Universitaria de Secuenciación Masiva y Bioinformática, Instituto de Biotecnología, Cuernavaca, México
- * E-mail: (ASF); (EM)
| | - Estrella Montero
- Laboratorio de Referencia e Investigación en Parasitología, Centro Nacional de Microbiología, ISCIII Majadahonda, Madrid, Spain
- * E-mail: (ASF); (EM)
| |
Collapse
|
29
|
Winkel BM, de Korne CM, van Oosterom MN, Staphorst D, Bunschoten A, Langenberg MC, Chevalley-Maurel SC, Janse CJ, Franke-Fayard B, van Leeuwen FW, Roestenberg M. A tracer-based method enables tracking of Plasmodium falciparum malaria parasites during human skin infection. Theranostics 2019; 9:2768-2778. [PMID: 31244921 PMCID: PMC6568182 DOI: 10.7150/thno.33467] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 02/22/2019] [Indexed: 11/10/2022] Open
Abstract
Introduction: The skin stage of malaria is a vital and vulnerable migratory life stage of the parasite. It has been characterised in rodent models, but remains wholly uninvestigated for human malaria parasites. To enable in depth analysis of not genetically modified (non-GMO) Plasmodium falciparum (Pf) sporozoite behaviour in human skin, we devised a labelling technology (Cy5M2, targeting the sporozoite mitochondrion) that supports tracking of individual non-GMO sporozoites in human skin. Methods: Sporozoite labelling with Cy5M2 was performed in vitro as well as via the feed of infected Anopheles mosquitos. Labelling was validated using confocal microscopy and flow cytometry and the fitness of labelled sporozoites was determined by analysis of infectivity to human hepatocytes in vitro, and in vivo in a rodent infection model. Using confocal video microscopy and custom software, single-sporozoite tracking studies in human skin-explants were performed. Results: Both in vitro and in mosquito labelling strategies yielded brightly fluorescent sporozoites of three different Plasmodium species. Cy5M2 uptake colocalized with MitoTracker® green and could be blocked using the known Translocator protein (TSPO)-inhibitor PK11195. This method supported the visualization and subsequent quantitative analysis of the migration patterns of individual non-GMO Pf sporozoites in human skin and did not affect the fitness of sporozoites. Conclusions: The ability to label and image non-GMO Plasmodium sporozoites provides the basis for detailed studies on the human skin stage of malaria with potential for in vivo translation. As such, it is an important tool for development of vaccines based on attenuated sporozoites and their route of administration.
Collapse
Affiliation(s)
- Béatrice M.F. Winkel
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Clarize M. de Korne
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Matthias N. van Oosterom
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Diego Staphorst
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Anton Bunschoten
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Laboratory of BioNanoTechnology Wageningen University and Research, Droevendaalsesteeg 4, 6708 PB Wageningen, The Netherlands
| | - Marijke C.C. Langenberg
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | | | - Chris J. Janse
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Blandine Franke-Fayard
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Fijs W.B. van Leeuwen
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Meta Roestenberg
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
30
|
Schloetel JG, Heine J, Cowman AF, Pasternak M. Guided STED nanoscopy enables super-resolution imaging of blood stage malaria parasites. Sci Rep 2019; 9:4674. [PMID: 30886187 PMCID: PMC6423018 DOI: 10.1038/s41598-019-40718-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 02/19/2019] [Indexed: 12/27/2022] Open
Abstract
Malaria remains a major burden world-wide, but the disease-causing parasites from the genus Plasmodium are difficult to study in vitro. Owing to the small size of the parasites, subcellular imaging poses a major challenge and the use of super-resolution techniques has been hindered by the parasites' sensitivity to light. This is particularly apparent during the blood-stage of the Plasmodium life cycle, which presents an important target for drug research. The iron-rich food vacuole of the parasite undergoes disintegration when illuminated with high-power lasers such as those required for high resolution in Stimulated Emission Depletion (STED) microscopy. This causes major damage to the sample precluding the use of this super-resolution technique. Here we present guided STED, a novel adaptive illumination (AI) STED approach, which takes advantage of the highly-reflective nature of the iron deposit in the cell to identify the most light-sensitive parts of the sample. Specifically in these parts, the high-power STED laser is deactivated automatically to prevent local damage. Guided STED nanoscopy finally allows super-resolution imaging of the whole Plasmodium life cycle, enabling multicolour imaging of blood-stage malaria parasites with resolutions down to 35 nm without sample destruction.
Collapse
Affiliation(s)
| | - Jörn Heine
- Abberior Instruments GmbH, 37077, Göttingen, Germany
| | - Alan F Cowman
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Michał Pasternak
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
31
|
Costa DM, Sá M, Teixeira AR, Loureiro I, Thouvenot C, Golba S, Amino R, Tavares J. TRSP is dispensable for the Plasmodium pre-erythrocytic phase. Sci Rep 2018; 8:15101. [PMID: 30305687 PMCID: PMC6180128 DOI: 10.1038/s41598-018-33398-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 09/26/2018] [Indexed: 11/09/2022] Open
Abstract
Plasmodium sporozoites deposited in the skin following a mosquito bite must migrate and invade blood vessels to complete their development in the liver. Once in the bloodstream, sporozoites arrest in the liver sinusoids, but the molecular determinants that mediate this specific homing are not yet genetically defined. Here we investigate the involvement of the thrombospondin-related sporozoite protein (TRSP) in this process using knockout Plasmodium berghei parasites and in vivo bioluminescence imaging in mice. Resorting to a homing assay, trsp knockout sporozoites were found to arrest in the liver similar to control parasites. Moreover, we found no defects in the establishment of infection in mice following inoculation of trsp knockout sporozoites via intravenous and cutaneous injection or mosquito bite. Accordingly, mutant sporozoites were also able to successfully invade hepatocytes in vitro. Altogether, these results suggest TRSP may have a redundant role in the completion of the pre-erythrocytic phase of the malaria parasite. Nonetheless, identifying molecules with paramount roles in this phase could aid in the search for new antigens needed for the design of a protective vaccine against malaria.
Collapse
Affiliation(s)
- David Mendes Costa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, 4200-135, Portugal
| | - Mónica Sá
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, 4200-135, Portugal
| | - Ana Rafaela Teixeira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, 4200-135, Portugal
| | - Inês Loureiro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, 4200-135, Portugal
| | - Catherine Thouvenot
- Center for Production and Infection of Anopheles, Institut Pasteur, Paris, 75015, France.,Ultrapole, Institut Pasteur, Paris, 75015, France
| | - Sylvain Golba
- Center for Production and Infection of Anopheles, Institut Pasteur, Paris, 75015, France
| | - Rogerio Amino
- Unit of Malaria Infection and Immunity, Institut Pasteur, Paris, 75015, France.
| | - Joana Tavares
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal. .,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, 4200-135, Portugal.
| |
Collapse
|
32
|
Glushakova S, Beck JR, Garten M, Busse BL, Nasamu AS, Tenkova-Heuser T, Heuser J, Goldberg DE, Zimmerberg J. Rounding precedes rupture and breakdown of vacuolar membranes minutes before malaria parasite egress from erythrocytes. Cell Microbiol 2018; 20:e12868. [PMID: 29900649 DOI: 10.1111/cmi.12868] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 05/25/2018] [Accepted: 06/05/2018] [Indexed: 01/17/2023]
Abstract
Because Plasmodium falciparum replicates inside of a parasitophorous vacuole (PV) within a human erythrocyte, parasite egress requires the rupture of two limiting membranes. Parasite Ca2+ , kinases, and proteases contribute to efficient egress; their coordination in space and time is not known. Here, the kinetics of parasite egress were linked to specific steps with specific compartment markers, using live-cell microscopy of parasites expressing PV-targeted fluorescent proteins, and specific egress inhibitors. Several minutes before egress, under control of parasite [Ca2+ ]i , the PV began rounding. Then after ~1.5 min, under control of PfPKG and SUB1, there was abrupt rupture of the PV membrane and release of vacuolar contents. Over the next ~6 min, there was progressive vacuolar membrane deterioration simultaneous with erythrocyte membrane distortion, lasting until the final minute of the egress programme when newly formed parasites mobilised and erythrocyte membranes permeabilised and then ruptured-a dramatic finale to the parasite cycle of replication.
Collapse
Affiliation(s)
- Svetlana Glushakova
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Josh R Beck
- Division of Infectious Diseases, Department of Medicine, Washington University, St. Louis, Missouri.,Department of Biomedical Sciences, Iowa State University, Ames, Iowa
| | - Matthias Garten
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Brad L Busse
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Armiyaw S Nasamu
- Division of Infectious Diseases, Department of Medicine, Washington University, St. Louis, Missouri
| | - Tatyana Tenkova-Heuser
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - John Heuser
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Daniel E Goldberg
- Division of Infectious Diseases, Department of Medicine, Washington University, St. Louis, Missouri
| | - Joshua Zimmerberg
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
33
|
Asada M, Hakimi H, Kawazu SI. The application of the HyPer fluorescent sensor in the real-time detection of H 2O 2 in Babesia bovis merozoites in vitro. Vet Parasitol 2018; 255:78-82. [PMID: 29773141 DOI: 10.1016/j.vetpar.2018.03.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/14/2018] [Accepted: 03/18/2018] [Indexed: 01/24/2023]
Abstract
In recent years, genetically encoded fluorescent probes have allowed a dramatic advancement in time-lapse imaging, enabling this imaging modality to be used to investigate intracellular events in several apicomplexan parasite species. In this study, we constructed a plasmid vector to stably express a genetically encoded H2O2 sensor probe called HyPer in Babesia bovis. The HyPer-transfected parasite population was successfully developed and subjected to a time-lapse imaging analysis under in vitro culture conditions. HyPer was capable of sensing an increasing H2O2 concentration in the parasite cells which was induced by the administration of paraquat as a superoxide donor. HyPer fluorescence co-staining with MitoTracker Red indicated the mitochondria as the major source of reactive oxygen species (ROS) in parasite cells. The fluctuating ROS dynamics in the parasite gliding toward, attaching to, and invading the target red blood cell was visualized and monitored in real time with the HyPer expressing parasite population. This is the first report to describe the application of the HyPer probe in an imaging analysis involving Babesia parasites. Hyper-expressing parasites can be widely utilized in studies to investigate the mechanisms of emergence and the reduction of oxidative stress, as well as the signal transduction in the parasite cells during host invasion and intercellular development.
Collapse
Affiliation(s)
- Masahito Asada
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto 1-12-4, Nagasaki, 852-8523, Japan; National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, 080-8555, Japan.
| | - Hassan Hakimi
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto 1-12-4, Nagasaki, 852-8523, Japan; National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, 080-8555, Japan.
| | - Shin-Ichiro Kawazu
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, 080-8555, Japan.
| |
Collapse
|
34
|
BioID Reveals Novel Proteins of the Plasmodium Parasitophorous Vacuole Membrane. mSphere 2018; 3:mSphere00522-17. [PMID: 29404413 PMCID: PMC5784244 DOI: 10.1128/msphere.00522-17] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 12/22/2017] [Indexed: 11/24/2022] Open
Abstract
Intracellular pathogens are often surrounded by a host-cell derived membrane. This membrane is modified by the pathogens to their own needs and is crucial for their intracellular lifestyle. In Plasmodium parasites, this membrane is referred to as the PVM and only a limited number of its proteins are known so far. Here, we applied in rodent P. berghei parasites a method called BioID, which is based on biotinylation of proximal and interacting proteins by the promiscuous biotin ligase BirA*, and demonstrated its usefulness in identification of novel PVM proteins. During their development within the vertebrate host, Plasmodium parasites infect hepatocytes and red blood cells. Within these cells, parasites are surrounded by a parasitophorous vacuole membrane (PVM). The PVM plays an essential role for the interaction of parasites with their host cells; however, only a limited number of proteins of this membrane have been identified so far. This is partially because systematic proteomic analysis of the protein content of the PVM has been difficult in the past, due to difficulties encountered in attempts to separate the PVM from other membranes such as the parasite plasma membrane. In this study, we adapted the BioID technique to in vitro-cultivated Plasmodium berghei blood stage parasites and utilized the promiscuous biotin ligase BirA* fused to PVM-resident exported protein 1 to biotinylate proteins of the PVM. These we further processed by affinity purification, liquid chromatography-tandem mass spectrometry (LC-MS/MS), and label-free quantitation, leading to a list of 61 known and candidate PVM proteins. Seven proteins were analyzed further during blood and liver stage development. This resulted in the identification of three novel PVM proteins, which were the serine/threonine protein phosphatase UIS2 (PlasmoDB accession no. PBANKA_1328000) and two conserved Plasmodium proteins with unknown functions (PBANKA_0519300 and PBANKA_0509000). In conclusion, our report expands the number of known PVM proteins and experimentally validates BioID as a powerful method to screen for novel constituents of specific cellular compartments in P. berghei. IMPORTANCE Intracellular pathogens are often surrounded by a host-cell derived membrane. This membrane is modified by the pathogens to their own needs and is crucial for their intracellular lifestyle. In Plasmodium parasites, this membrane is referred to as the PVM and only a limited number of its proteins are known so far. Here, we applied in rodent P. berghei parasites a method called BioID, which is based on biotinylation of proximal and interacting proteins by the promiscuous biotin ligase BirA*, and demonstrated its usefulness in identification of novel PVM proteins.
Collapse
|
35
|
Zuck M, Austin LS, Danziger SA, Aitchison JD, Kaushansky A. The Promise of Systems Biology Approaches for Revealing Host Pathogen Interactions in Malaria. Front Microbiol 2017; 8:2183. [PMID: 29201016 PMCID: PMC5696578 DOI: 10.3389/fmicb.2017.02183] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 10/24/2017] [Indexed: 12/18/2022] Open
Abstract
Despite global eradication efforts over the past century, malaria remains a devastating public health burden, causing almost half a million deaths annually (WHO, 2016). A detailed understanding of the mechanisms that control malaria infection has been hindered by technical challenges of studying a complex parasite life cycle in multiple hosts. While many interventions targeting the parasite have been implemented, the complex biology of Plasmodium poses a major challenge, and must be addressed to enable eradication. New approaches for elucidating key host-parasite interactions, and predicting how the parasite will respond in a variety of biological settings, could dramatically enhance the efficacy and longevity of intervention strategies. The field of systems biology has developed methodologies and principles that are well poised to meet these challenges. In this review, we focus our attention on the Liver Stage of the Plasmodium lifecycle and issue a “call to arms” for using systems biology approaches to forge a new era in malaria research. These approaches will reveal insights into the complex interplay between host and pathogen, and could ultimately lead to novel intervention strategies that contribute to malaria eradication.
Collapse
Affiliation(s)
- Meghan Zuck
- Center for Infectious Disease Research, formerly Seattle Biomedical Research Institute, Seattle, WA, United States
| | - Laura S Austin
- Center for Infectious Disease Research, formerly Seattle Biomedical Research Institute, Seattle, WA, United States
| | - Samuel A Danziger
- Center for Infectious Disease Research, formerly Seattle Biomedical Research Institute, Seattle, WA, United States.,Institute for Systems Biology, Seattle, WA, United States
| | - John D Aitchison
- Center for Infectious Disease Research, formerly Seattle Biomedical Research Institute, Seattle, WA, United States.,Institute for Systems Biology, Seattle, WA, United States
| | - Alexis Kaushansky
- Center for Infectious Disease Research, formerly Seattle Biomedical Research Institute, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States
| |
Collapse
|
36
|
Phospholipases during membrane dynamics in malaria parasites. Int J Med Microbiol 2017; 308:129-141. [PMID: 28988696 DOI: 10.1016/j.ijmm.2017.09.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/15/2017] [Accepted: 09/19/2017] [Indexed: 12/26/2022] Open
Abstract
Plasmodium parasites, the causative agents of malaria, display a well-regulated lipid metabolism required to ensure their survival in the human host as well as in the mosquito vector. The fine-tuning of lipid metabolic pathways is particularly important for the parasites during the rapid erythrocytic infection cycles, and thus enzymes involved in lipid metabolic processes represent prime targets for malaria chemotherapeutics. While plasmodial enzymes involved in lipid synthesis and acquisition have been studied in the past, to date not much is known about the roles of phospholipases for proliferation and transmission of the malaria parasite. These phospholipid-hydrolyzing esterases are crucial for membrane dynamics during host cell infection and egress by the parasite as well as for replication and cell signaling, and thus they are considered important virulence factors. In this review, we provide a comprehensive bioinformatic analysis of plasmodial phospholipases identified to date. We further summarize previous findings on the lipid metabolism of Plasmodium, highlight the roles of phospholipases during parasite life-cycle progression, and discuss the plasmodial phospholipases as potential targets for malaria therapy.
Collapse
|
37
|
|
38
|
A Plasmodium plasma membrane reporter reveals membrane dynamics by live-cell microscopy. Sci Rep 2017; 7:9740. [PMID: 28851956 PMCID: PMC5575152 DOI: 10.1038/s41598-017-09569-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 07/26/2017] [Indexed: 11/08/2022] Open
Abstract
During asexual replication within the Anopheles mosquito and their vertebrate host, Plasmodium parasites depend on the generation of a massive amount of new plasma membrane to produce thousands of daughter parasites. How the parasite plasma membrane (PPM) is formed has mostly been studied by electron microscopy, which does not allow an insight into the dynamics of this process. We generated a Plasmodium berghei reporter parasite line by GFP-tagging of a non-essential PPM-localized protein, and followed plasma membrane development in living parasites through the entire Plasmodium life cycle. By generating double-fluorescent parasites in which the PPM is visualized in combination with the parasite endoplasmic reticulum, we show that membrane contact sites are formed between both membrane systems during oocyst and liver stage development that might be used to deliver lipids to the dramatically expanding PPM. In conclusion, we have established a powerful tool to follow PPM development in living parasites, which promises to greatly expand our knowledge of membrane biology in the Plasmodium parasite.
Collapse
|
39
|
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
|
40
|
Agop-Nersesian C, De Niz M, Niklaus L, Prado M, Eickel N, Heussler VT. Shedding of host autophagic proteins from the parasitophorous vacuolar membrane of Plasmodium berghei. Sci Rep 2017; 7:2191. [PMID: 28526861 PMCID: PMC5438358 DOI: 10.1038/s41598-017-02156-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 04/05/2017] [Indexed: 01/05/2023] Open
Abstract
The hepatic stage of the malaria parasite Plasmodium is accompanied by an autophagy-mediated host response directly targeting the parasitophorous vacuolar membrane (PVM) harbouring the parasite. Removal of the PVM-associated autophagic proteins such as ubiquitin, p62, and LC3 correlates with parasite survival. Yet, it is unclear how Plasmodium avoids the deleterious effects of selective autophagy. Here we show that parasites trap host autophagic factors in the tubovesicular network (TVN), an expansion of the PVM into the host cytoplasm. In proliferating parasites, PVM-associated LC3 becomes immediately redirected into the TVN, where it accumulates distally from the parasite’s replicative centre. Finally, the host factors are shed as vesicles into the host cytoplasm. This strategy may enable the parasite to balance the benefits of the enhanced host catabolic activity with the risk of being eliminated by the cell’s cytosolic immune defence.
Collapse
Affiliation(s)
- Carolina Agop-Nersesian
- Institute of Cell Biology, University of Bern, 3012, Bern, Switzerland. .,Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, MA, 02118, USA.
| | - Mariana De Niz
- Institute of Cell Biology, University of Bern, 3012, Bern, Switzerland.,Wellcome Centre for Molecular Parasitology, University of Glasgow, G12 8QQ, Glasgow, UK
| | - Livia Niklaus
- Institute of Cell Biology, University of Bern, 3012, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012, Bern, Switzerland
| | - Monica Prado
- Bernhard Nocht Institute of Tropical Medicine, 20359, Hamburg, Germany.,Centro de Investigación en Enfermedades Tropicales (CIET), Universidad de Costa Rica, San José, Costa Rica, USA
| | - Nina Eickel
- Institute of Cell Biology, University of Bern, 3012, Bern, Switzerland.,CSL Behring, Bern, Switzerland
| | - Volker T Heussler
- Institute of Cell Biology, University of Bern, 3012, Bern, Switzerland.
| |
Collapse
|
41
|
Tavares J, Costa DM, Teixeira AR, Cordeiro-da-Silva A, Amino R. In vivo imaging of pathogen homing to the host tissues. Methods 2017; 127:37-44. [PMID: 28522323 DOI: 10.1016/j.ymeth.2017.05.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 04/19/2017] [Accepted: 05/10/2017] [Indexed: 12/19/2022] Open
Abstract
Hematogenous dissemination followed by tissue tropism is a characteristic of the infectious process of many pathogens including those transmitted by blood-feeding vectors. After entering into the blood circulation, these pathogens must arrest in the target organ before they infect a specific tissue. Here, we describe a non-invasive method to visualize and quantify the homing of pathogens to the host tissues. By using in vivo bioluminescence imaging we quantify the accumulation of luciferase-expressing parasites in the host organs during the first minutes following their intravascular inoculation in mice. Using this technique we show that in the malarial infection, once in the blood circulation, most of bioluminescent Plasmodium berghei sporozoites, the parasite stage transmitted to the host skin by a mosquito bite, rapidly home to the liver where they invade and develop inside hepatocytes. This homing is specific to this developmental stage since blood stage parasites do not accumulate in the liver, as well as extracellular Trypanosoma brucei bloodstream forms and liver-infecting Leishmania infantum amastigotes. Finally, this method can be used to study the dynamics of tissue tropism of parasites, dissect the molecular and cellular basis of their increased arrest in organs and to evaluate immune interventions designed to block this targeted interaction.
Collapse
Affiliation(s)
- Joana Tavares
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Portugal.
| | - David Mendes Costa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Portugal
| | - Ana Rafaela Teixeira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Portugal
| | - Anabela Cordeiro-da-Silva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Portugal; Faculdade de Farmácia da Universidade do Porto, Departamento de Ciências Biológicas, Portugal
| | - Rogerio Amino
- Unit of Malaria Infection and Immunity, Institut Pasteur, Paris, France.
| |
Collapse
|
42
|
Klug D, Frischknecht F. Motility precedes egress of malaria parasites from oocysts. eLife 2017; 6. [PMID: 28115054 PMCID: PMC5262382 DOI: 10.7554/elife.19157] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 12/16/2016] [Indexed: 12/19/2022] Open
Abstract
Malaria is transmitted when an infected Anopheles mosquito deposits Plasmodium sporozoites in the skin during a bite. Sporozoites are formed within oocysts at the mosquito midgut wall and are released into the hemolymph, from where they invade the salivary glands and are subsequently transmitted to the vertebrate host. We found that a thrombospondin-repeat containing sporozoite-specific protein named thrombospondin-releated protein 1 (TRP1) is important for oocyst egress and salivary gland invasion, and hence for the transmission of malaria. We imaged the release of sporozoites from oocysts in situ, which was preceded by active motility. Parasites lacking TRP1 failed to migrate within oocysts and did not egress, suggesting that TRP1 is a vital component of the events that precede intra-oocyst motility and subsequently sporozoite egress and salivary gland invasion. DOI:http://dx.doi.org/10.7554/eLife.19157.001 Malaria is caused by a parasite transmitted by certain types of mosquito. The parasite lives in different organs within its vertebrate animal and insect hosts and to cope with these different environments it has a complex life cycle with several highly specialized life stages. To move from an infected mosquito into vertebrates the parasite produces spore-like cells called sporozoites that are able to enter different tissues and move very fast. These cells develop inside parasite-made structures called oocysts, which form at the stomach wall of the mosquito. After emerging from the oocyst, sporozoites float through the mosquito’s circulatory system and eventually enter the salivary glands where they can be transmitted to vertebrates when the mosquito bites. Efforts to develop malaria treatments and vaccines have focused on understanding the parasite’s life cycle and identifying ways to control or eradicate key stages. Most researchers focus on the stage where the parasite is living in the vertebrate and actively causing disease, while the events in the mosquito are less intensely investigated. While several parasite proteins have been shown to be important for the release of sporozoites from oocysts, the molecular events leading to this release have not yet been fully resolved. Klug and Frischknecht used time-lapse microscopy to film the release of the sporozoites of a malaria parasite known as Plasmodium berghei. The experiments show that the sporozoites can leave oocysts in several different ways. Furthermore, Klug and Frischknecht identified a new parasite protein named TRP1 that is essential for the sporozoites to leave oocysts and invade the salivary glands. Sporozoites lacking TRP1 were not able to move and they were unable to leave the oocyst or invade the salivary glands. Klug and Frischknecht propose a new working model of the molecular events that govern sporozoite release in which TRP1 is required for sporozoites to move prior to their exit from oocysts. In the future, using the same techniques to analyze genetically modified parasites will help to reveal more details about sporozoite release. DOI:http://dx.doi.org/10.7554/eLife.19157.002
Collapse
Affiliation(s)
- Dennis Klug
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| |
Collapse
|