101
|
Collins CR, Hackett F, Atid J, Tan MSY, Blackman MJ. The Plasmodium falciparum pseudoprotease SERA5 regulates the kinetics and efficiency of malaria parasite egress from host erythrocytes. PLoS Pathog 2017; 13:e1006453. [PMID: 28683142 PMCID: PMC5500368 DOI: 10.1371/journal.ppat.1006453] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 06/07/2017] [Indexed: 02/06/2023] Open
Abstract
Egress of the malaria parasite Plasmodium falciparum from its host red blood cell is a rapid, highly regulated event that is essential for maintenance and completion of the parasite life cycle. Egress is protease-dependent and is temporally associated with extensive proteolytic modification of parasite proteins, including a family of papain-like proteins called SERA that are expressed in the parasite parasitophorous vacuole. Previous work has shown that the most abundant SERA, SERA5, plays an important but non-enzymatic role in asexual blood stages. SERA5 is extensively proteolytically processed by a parasite serine protease called SUB1 as well as an unidentified cysteine protease just prior to egress. However, neither the function of SERA5 nor the role of its processing is known. Here we show that conditional disruption of the SERA5 gene, or of both the SERA5 and related SERA4 genes simultaneously, results in a dramatic egress and replication defect characterised by premature host cell rupture and the failure of daughter merozoites to efficiently disseminate, instead being transiently retained within residual bounding membranes. SERA5 is not required for poration (permeabilization) or vesiculation of the host cell membrane at egress, but the premature rupture phenotype requires the activity of a parasite or host cell cysteine protease. Complementation of SERA5 null parasites by ectopic expression of wild-type SERA5 reversed the egress defect, whereas expression of a SERA5 mutant refractory to processing failed to rescue the phenotype. Our findings implicate SERA5 as an important regulator of the kinetics and efficiency of egress and suggest that proteolytic modification is required for SERA5 function. In addition, our study reveals that efficient egress requires tight control of the timing of membrane rupture. Malaria, a disease that kills hundreds of thousands of people each year, is caused by a single-celled parasite that grows in red blood cells of infected individuals. Following each round of parasite multiplication, the infected red cells are actively ruptured in a process called egress, releasing a new generation of parasites. Egress is essential for progression to clinical disease, but little is known about how it is controlled. In this work we set out to address the function in egress of a Plasmodium falciparum protein called SERA5, an abundant component of the vacuole in which the parasite grows. We show that parasites lacking SERA5 (or lacking both SERA5 and a closely-related protein called SERA4) undergo accelerated but defective egress in which the bounding vacuole and red cell membranes do not rupture properly. This impedes the escape and subsequent replication of the newly-developed parasites. We also show that modification of SERA5 by parasites proteases just prior to egress is important for SERA5 function. Our results show that SERA5 is a ‘negative regulator’ of egress, controlling the speed of the pathway that leads to disruption of the membranes surrounding the intracellular parasite. Our findings increase our understanding of the molecular mechanisms underlying malarial egress and show that efficient egress requires tight control of the timing of membrane rupture.
Collapse
Affiliation(s)
- Christine R. Collins
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Fiona Hackett
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Jonathan Atid
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Michele Ser Ying Tan
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Michael J. Blackman
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
- Department of Pathogen Molecular Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
- * E-mail:
| |
Collapse
|
102
|
Knuepfer E, Napiorkowska M, van Ooij C, Holder AA. Generating conditional gene knockouts in Plasmodium - a toolkit to produce stable DiCre recombinase-expressing parasite lines using CRISPR/Cas9. Sci Rep 2017. [PMID: 28634346 PMCID: PMC5478596 DOI: 10.1038/s41598-017-03984-3] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Successful establishment of CRISPR/Cas9 genome editing technology in Plasmodium spp. has provided a powerful tool to transform Plasmodium falciparum into a genetically more tractable organism. Conditional gene regulation approaches are required to study the function of gene products critical for growth and erythrocyte invasion of blood stage parasites. Here we employ CRISPR/Cas9 to facilitate use of the dimerisable Cre-recombinase (DiCre) that is frequently used to mediate the excision and loss of loxP-flanked DNA sequences in a rapamycin controlled manner. We describe novel CRISPR/Cas9 transfection plasmids and approaches for the speedy, stable and marker-free introduction of transgenes encoding the DiCre recombinase into genomic loci dispensable for blood stage development. Together these plasmids form a toolkit that will allow the rapid generation of transgenic DiCre-expressing P. falciparum lines in any genetic background. Furthermore, the newly developed 3D7-derived parasite lines, constitutively and stably expressing DiCre, generated using this toolkit will prove useful for the analysis of gene products. Lastly, we introduce an improved treatment protocol that uses a lower rapamycin concentration and shorter treatment times, leading to loxP-guided recombination with close to 100% efficiency within the same replication cycle.
Collapse
Affiliation(s)
- Ellen Knuepfer
- Malaria Parasitology Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom.
| | - Marta Napiorkowska
- Malaria Biochemistry Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom.,Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, 4058, Switzerland
| | - Christiaan van Ooij
- Malaria Biochemistry Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom.
| | - Anthony A Holder
- Malaria Parasitology Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom.
| |
Collapse
|
103
|
Hale VL, Watermeyer JM, Hackett F, Vizcay-Barrena G, van Ooij C, Thomas JA, Spink MC, Harkiolaki M, Duke E, Fleck RA, Blackman MJ, Saibil HR. Parasitophorous vacuole poration precedes its rupture and rapid host erythrocyte cytoskeleton collapse in Plasmodium falciparum egress. Proc Natl Acad Sci U S A 2017; 114:3439-3444. [PMID: 28292906 PMCID: PMC5380091 DOI: 10.1073/pnas.1619441114] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In the asexual blood stages of malarial infection, merozoites invade erythrocytes and replicate within a parasitophorous vacuole to form daughter cells that eventually exit (egress) by sequential rupture of the vacuole and erythrocyte membranes. The current model is that PKG, a malarial cGMP-dependent protein kinase, triggers egress, activating malarial proteases and other effectors. Using selective inhibitors of either PKG or cysteine proteases to separately inhibit the sequential steps in membrane perforation, combined with video microscopy, electron tomography, electron energy loss spectroscopy, and soft X-ray tomography of mature intracellular Plasmodium falciparum parasites, we resolve intermediate steps in egress. We show that the parasitophorous vacuole membrane (PVM) is permeabilized 10-30 min before its PKG-triggered breakdown into multilayered vesicles. Just before PVM breakdown, the host red cell undergoes an abrupt, dramatic shape change due to the sudden breakdown of the erythrocyte cytoskeleton, before permeabilization and eventual rupture of the erythrocyte membrane to release the parasites. In contrast to the previous view of PKG-triggered initiation of egress and a gradual dismantling of the host erythrocyte cytoskeleton over the course of schizont development, our findings identify an initial step in egress and show that host cell cytoskeleton breakdown is restricted to a narrow time window within the final stages of egress.
Collapse
Affiliation(s)
- Victoria L Hale
- Crystallography, Institute of Structural and Molecular Biology, Birkbeck College, London, WC1E 7HX, United Kingdom
| | - Jean M Watermeyer
- Crystallography, Institute of Structural and Molecular Biology, Birkbeck College, London, WC1E 7HX, United Kingdom
| | - Fiona Hackett
- Francis Crick Institute, London, NW1 1AT, United Kingdom
| | - Gema Vizcay-Barrena
- Centre for Ultrastructural Imaging, Kings College London, London, SE1 9RT, United Kingdom
| | | | - James A Thomas
- Francis Crick Institute, London, NW1 1AT, United Kingdom
| | | | | | | | - Roland A Fleck
- Centre for Ultrastructural Imaging, Kings College London, London, SE1 9RT, United Kingdom
| | - Michael J Blackman
- Francis Crick Institute, London, NW1 1AT, United Kingdom
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, United Kingdom
| | - Helen R Saibil
- Crystallography, Institute of Structural and Molecular Biology, Birkbeck College, London, WC1E 7HX, United Kingdom;
| |
Collapse
|
104
|
Kariuki SM, Selhorst P, Ariën KK, Dorfman JR. The HIV-1 transmission bottleneck. Retrovirology 2017; 14:22. [PMID: 28335782 PMCID: PMC5364581 DOI: 10.1186/s12977-017-0343-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 03/05/2017] [Indexed: 02/07/2023] Open
Abstract
It is well established that most new systemic infections of HIV-1 can be traced back to one or a limited number of founder viruses. Usually, these founders are more closely related to minor HIV-1 populations in the blood of the presumed donor than to more abundant lineages. This has led to the widely accepted idea that transmission selects for viral characteristics that facilitate crossing the mucosal barrier of the recipient’s genital tract, although the specific selective forces or advantages are not completely defined. However, there are other steps along the way to becoming a founder virus at which selection may occur. These steps include the transition from the donor’s general circulation to the genital tract compartment, survival within the transmission fluid, and establishment of a nascent stable local infection in the recipient’s genital tract. Finally, there is the possibility that important narrowing events may also occur during establishment of systemic infection. This is suggested by the surprising observation that the number of founder viruses detected after transmission in intravenous drug users is also limited. Although some of these steps may be heavily selective, others may result mostly in a stochastic narrowing of the available founder pool. Collectively, they shape the initial infection in each recipient.
Collapse
Affiliation(s)
- Samuel Mundia Kariuki
- Division of Immunology, Department of Pathology, Falmouth 3.25, University of Cape Town, Anzio Rd, Observatory, Cape Town, 7925, South Africa.,International Centre for Genetic Engineering and Biotechnology, Cape Town, South Africa.,Department of Biological Sciences, University of Eldoret, Eldoret, Kenya
| | - Philippe Selhorst
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Kevin K Ariën
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium.,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jeffrey R Dorfman
- Division of Immunology, Department of Pathology, Falmouth 3.25, University of Cape Town, Anzio Rd, Observatory, Cape Town, 7925, South Africa.
| |
Collapse
|
105
|
Birnbaum J, Flemming S, Reichard N, Soares AB, Mesén-Ramírez P, Jonscher E, Bergmann B, Spielmann T. A genetic system to study Plasmodium falciparum protein function. Nat Methods 2017; 14:450-456. [DOI: 10.1038/nmeth.4223] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 02/06/2017] [Indexed: 02/07/2023]
|
106
|
Sherling ES, Knuepfer E, Brzostowski JA, Miller LH, Blackman MJ, van Ooij C. The Plasmodium falciparum rhoptry protein RhopH3 plays essential roles in host cell invasion and nutrient uptake. eLife 2017; 6. [PMID: 28252384 PMCID: PMC5365315 DOI: 10.7554/elife.23239] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 02/26/2017] [Indexed: 11/18/2022] Open
Abstract
Merozoites of the protozoan parasite responsible for the most virulent form of malaria, Plasmodium falciparum, invade erythrocytes. Invasion involves discharge of rhoptries, specialized secretory organelles. Once intracellular, parasites induce increased nutrient uptake by generating new permeability pathways (NPP) including a Plasmodium surface anion channel (PSAC). RhopH1/Clag3, one member of the three-protein RhopH complex, is important for PSAC/NPP activity. However, the roles of the other members of the RhopH complex in PSAC/NPP establishment are unknown and it is unclear whether any of the RhopH proteins play a role in invasion. Here we demonstrate that RhopH3, the smallest component of the complex, is essential for parasite survival. Conditional truncation of RhopH3 substantially reduces invasive capacity. Those mutant parasites that do invade are defective in nutrient import and die. Our results identify a dual role for RhopH3 that links erythrocyte invasion to formation of the PSAC/NPP essential for parasite survival within host erythrocytes. DOI:http://dx.doi.org/10.7554/eLife.23239.001 Malaria is a life-threatening disease that affects millions of people around the world. The parasites that cause malaria have a complex life cycle that involves infecting both mosquitoes and mammals, including humans. In humans, the parasites spend part of their life cycle inside red blood cells, which causes the symptoms of the disease. In order to survive and multiply, malaria parasites need to make the red blood cell more permeable so that it can absorb nutrients from the blood stream and get rid of the toxic waste products they generate. It remains unclear how the parasites do this, but previous research has shown that the parasites produce channel-like proteins that make red blood cells more permeable to nutrients. One of the proteins involved in this process forms part of a complex with two other proteins, called RhopH2 and RhopH3. It is not known what these other two proteins do, and whether they are necessary for creating the new nutrient channels. Sherling et al. studied the RhopH3 protein to see if it is required to make red blood cells more permeable. The experiments used a genetically modified version of the parasite, in which RhopH3 no longer interacted with the two other proteins. The findings show that RhopH3 has two important roles: first, parasites need it to invade the red blood cells, and second, parasites cannot get nutrients into the red blood cell without RhopH3. Most antimalarial drugs work by preventing parasite replication in red blood cells, but parasites are becoming increasingly resistant to these drugs. Understanding which proteins allow parasites to invade and grow within blood cells will further the development of new malaria medication. The next step will be to understand the molecular mechanisms by which RhopH3 promotes invasion and subsequently facilitates nutrient uptake, and will help researchers to explore its potential as a drug target. DOI:http://dx.doi.org/10.7554/eLife.23239.002
Collapse
Affiliation(s)
- Emma S Sherling
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom.,Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, United States
| | - Ellen Knuepfer
- Malaria Parasitology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Joseph A Brzostowski
- Laboratory of Immunogenetics Imaging Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, United States
| | - Louis H Miller
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, United States
| | - Michael J Blackman
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom.,Department of Pathogen Molecular Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Christiaan van Ooij
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
107
|
Tools for attenuation of gene expression in malaria parasites. Int J Parasitol 2017; 47:385-398. [PMID: 28153780 DOI: 10.1016/j.ijpara.2016.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/15/2016] [Accepted: 11/28/2016] [Indexed: 12/30/2022]
Abstract
An understanding of the biology of Plasmodium parasites, which are the causative agents of the disease malaria, requires study of gene function. Various reverse genetic tools have been described for determining gene function. These tools can be broadly grouped as trans- and cis-acting. Trans-acting tools control gene functions through synthetic nucleic acid probe molecules matching the sequence of the gene of interest. Once delivered to the parasite, the probe engages with the mRNA of the target gene and attenuates its function. Cis-acting tools control gene function through elements introduced into the gene of interest by DNA transfection. The expression of the modified gene can be controlled using external agents, typically small molecule ligands. In this review, we discuss the strengths and weaknesses of these tools to guide researchers in selecting the appropriate tool for studies of gene function, and for guiding future refinements of these tools.
Collapse
|
108
|
Hill RJ, Ringel A, Knuepfer E, Moon RW, Blackman MJ, van Ooij C. Regulation and Essentiality of the StAR-related Lipid Transfer (START) Domain-containing Phospholipid Transfer Protein PFA0210c in Malaria Parasites. J Biol Chem 2016; 291:24280-24292. [PMID: 27694132 PMCID: PMC5104948 DOI: 10.1074/jbc.m116.740506] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 09/23/2016] [Indexed: 12/22/2022] Open
Abstract
StAR-related lipid transfer (START) domains are phospholipid- or sterol-binding modules that are present in many proteins. START domain-containing proteins (START proteins) play important functions in eukaryotic cells, including the redistribution of phospholipids to subcellular compartments and delivering sterols to the mitochondrion for steroid synthesis. How the activity of the START domain is regulated remains unknown for most of these proteins. The Plasmodium falciparum START protein PFA0210c (PF3D7_0104200) is a broad-spectrum phospholipid transfer protein that is conserved in all sequenced Plasmodium species and is most closely related to the mammalian START proteins STARD2 and STARD7. PFA0210c is unusual in that it contains a signal sequence and a PEXEL export motif that together mediate transfer of the protein from the parasite to the host erythrocyte. The protein also contains a C-terminal extension, which is very uncommon among mammalian START proteins. Whereas the biochemical properties of PFA0210c have been characterized, the function of the protein remains unknown. Here, we provide evidence that the unusual C-terminal extension negatively regulates phospholipid transfer activity. Furthermore, we use the genetically tractable Plasmodium knowlesi model and recently developed genetic technology in P. falciparum to show that the protein is essential for growth of the parasite during the clinically relevant asexual blood stage life cycle. Finally, we show that the regulation of phospholipid transfer by PFA0210c is required in vivo, and we identify a potential second regulatory domain. These findings provide insight into a novel mechanism of regulation of phospholipid transfer in vivo and may have important implications for the interaction of the malaria parasite with its host cell.
Collapse
Affiliation(s)
- Ross J Hill
- From the The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London NW7 1AA and
| | - Alessa Ringel
- From the The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London NW7 1AA and
| | - Ellen Knuepfer
- From the The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London NW7 1AA and
| | | | - Michael J Blackman
- From the The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London NW7 1AA and
- Pathogen Molecular Biology, London School of Hygiene & Tropical Medicine, London WC1E 7HT, United Kingdom
| | - Christiaan van Ooij
- From the The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London NW7 1AA and
| |
Collapse
|
109
|
Singer M, Frischknecht F. Time for Genome Editing: Next-Generation Attenuated Malaria Parasites. Trends Parasitol 2016; 33:202-213. [PMID: 27793562 DOI: 10.1016/j.pt.2016.09.012] [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: 07/15/2016] [Revised: 09/20/2016] [Accepted: 09/26/2016] [Indexed: 12/20/2022]
Abstract
Immunization with malaria parasites that developmentally arrest in or immediately after the liver stage is the only way currently known to confer sterilizing immunity in both humans and rodent models. There are various ways to attenuate parasite development resulting in different timings of arrest, which has a significant impact on vaccination efficiency. To understand what most impacts vaccination efficiency, newly developed gain-of-function methods can now be used to generate a wide array of differently attenuated parasites. The combination of multiple attenuation approaches offers the potential to engineer efficiently attenuated Plasmodium parasites and learn about their fascinating biology at the same time. Here we discuss recent studies and the potential of targeted parasite manipulation using genome editing to develop live attenuated malaria vaccines.
Collapse
Affiliation(s)
- Mirko Singer
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany.
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany.
| |
Collapse
|
110
|
A Telomeric Cluster of Antimony Resistance Genes on Chromosome 34 of Leishmania infantum. Antimicrob Agents Chemother 2016; 60:5262-75. [PMID: 27324767 DOI: 10.1128/aac.00544-16] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/11/2016] [Indexed: 11/20/2022] Open
Abstract
The mechanisms underlying the drug resistance of Leishmania spp. are manifold and not completely identified. Apart from the highly conserved multidrug resistance gene family known from higher eukaryotes, Leishmania spp. also possess genus-specific resistance marker genes. One of them, ARM58, was first identified in Leishmania braziliensis using a functional cloning approach, and its domain structure was characterized in L. infantum Here we report that L. infantum ARM58 is part of a gene cluster at the telomeric end of chromosome 34 also comprising the neighboring genes ARM56 and HSP23. We show that overexpression of all three genes can confer antimony resistance to intracellular amastigotes. Upon overexpression in L. donovani, ARM58 and ARM56 are secreted via exosomes, suggesting a scavenger/secretion mechanism of action. Using a combination of functional cloning and next-generation sequencing, we found that the gene cluster was selected only under antimonyl tartrate challenge and weakly under Cu(2+) challenge but not under sodium arsenite, Cd(2+), or miltefosine challenge. The selective advantage is less pronounced in intracellular amastigotes treated with the sodium stibogluconate, possibly due to the known macrophage-stimulatory activity of this drug, against which these resistance markers may not be active. Our data point to the specificity of these three genes for antimony resistance.
Collapse
|
111
|
Das S, Hertrich N, Perrin AJ, Withers-Martinez C, Collins CR, Jones ML, Watermeyer JM, Fobes ET, Martin SR, Saibil HR, Wright GJ, Treeck M, Epp C, Blackman MJ. Processing of Plasmodium falciparum Merozoite Surface Protein MSP1 Activates a Spectrin-Binding Function Enabling Parasite Egress from RBCs. Cell Host Microbe 2016; 18:433-44. [PMID: 26468747 PMCID: PMC4608996 DOI: 10.1016/j.chom.2015.09.007] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 09/17/2015] [Accepted: 09/18/2015] [Indexed: 11/09/2022]
Abstract
The malaria parasite Plasmodium falciparum replicates within erythrocytes, producing progeny merozoites that are released from infected cells via a poorly understood process called egress. The most abundant merozoite surface protein, MSP1, is synthesized as a large precursor that undergoes proteolytic maturation by the parasite protease SUB1 just prior to egress. The function of MSP1 and its processing are unknown. Here we show that SUB1-mediated processing of MSP1 is important for parasite viability. Processing modifies the secondary structure of MSP1 and activates its capacity to bind spectrin, a molecular scaffold protein that is the major component of the host erythrocyte cytoskeleton. Parasites expressing an inefficiently processed MSP1 mutant show delayed egress, and merozoites lacking surface-bound MSP1 display a severe egress defect. Our results indicate that interactions between SUB1-processed merozoite surface MSP1 and the spectrin network of the erythrocyte cytoskeleton facilitate host erythrocyte rupture to enable parasite egress. Merozoite surface protein MSP1 processing is important for P. falciparum viability Proteolytic processing activates MSP1’s heparin and spectrin-binding functions The rate of MSP1 processing governs the kinetics of parasite egress Loss of parasite surface MSP1 results in a severe egress defect
Collapse
Affiliation(s)
- Sujaan Das
- The Francis Crick Institute, Mill Hill Laboratory, Mill Hill, London, NW7 1AA, UK
| | - Nadine Hertrich
- Department für Infektiologie, Parasitologie, Universitätsklinikum Heidelberg, D-69120 Heidelberg, Germany
| | - Abigail J Perrin
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1HH, UK
| | | | - Christine R Collins
- The Francis Crick Institute, Mill Hill Laboratory, Mill Hill, London, NW7 1AA, UK
| | - Matthew L Jones
- The Francis Crick Institute, Mill Hill Laboratory, Mill Hill, London, NW7 1AA, UK
| | - Jean M Watermeyer
- Department of Crystallography, Birkbeck College, London, WC1E 7HX, UK
| | - Elmar T Fobes
- Department für Infektiologie, Parasitologie, Universitätsklinikum Heidelberg, D-69120 Heidelberg, Germany
| | - Stephen R Martin
- The Francis Crick Institute, Mill Hill Laboratory, Mill Hill, London, NW7 1AA, UK
| | - Helen R Saibil
- Department of Crystallography, Birkbeck College, London, WC1E 7HX, UK
| | - Gavin J Wright
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1HH, UK
| | - Moritz Treeck
- The Francis Crick Institute, Mill Hill Laboratory, Mill Hill, London, NW7 1AA, UK
| | - Christian Epp
- Department für Infektiologie, Parasitologie, Universitätsklinikum Heidelberg, D-69120 Heidelberg, Germany
| | - Michael J Blackman
- The Francis Crick Institute, Mill Hill Laboratory, Mill Hill, London, NW7 1AA, UK; Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK.
| |
Collapse
|
112
|
Essential Role of the PfRh5/PfRipr/CyRPA Complex during Plasmodium falciparum Invasion of Erythrocytes. Cell Host Microbe 2016; 20:60-71. [DOI: 10.1016/j.chom.2016.06.004] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 04/05/2016] [Accepted: 05/18/2016] [Indexed: 01/30/2023]
|
113
|
Development and Application of a Simple Plaque Assay for the Human Malaria Parasite Plasmodium falciparum. PLoS One 2016; 11:e0157873. [PMID: 27332706 PMCID: PMC4917082 DOI: 10.1371/journal.pone.0157873] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 06/06/2016] [Indexed: 02/03/2023] Open
Abstract
Malaria is caused by an obligate intracellular protozoan parasite that replicates within and destroys erythrocytes. Asexual blood stages of the causative agent of the most virulent form of human malaria, Plasmodium falciparum, can be cultivated indefinitely in vitro in human erythrocytes, facilitating experimental analysis of parasite cell biology, biochemistry and genetics. However, efforts to improve understanding of the basic biology of this important pathogen and to develop urgently required new antimalarial drugs and vaccines, suffer from a paucity of basic research tools. This includes a simple means of quantifying the effects of drugs, antibodies and gene modifications on parasite fitness and replication rates. Here we describe the development and validation of an extremely simple, robust plaque assay that can be used to visualise parasite replication and resulting host erythrocyte destruction at the level of clonal parasite populations. We demonstrate applications of the plaque assay by using it for the phenotypic characterisation of two P. falciparum conditional mutants displaying reduced fitness in vitro.
Collapse
|
114
|
Späth GF, Clos J. Joining forces: first application of a rapamycin-induced dimerizable Cre system for conditional null mutant analysis in Leishmania. Mol Microbiol 2016; 100:923-7. [PMID: 26991431 DOI: 10.1111/mmi.13374] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2016] [Indexed: 11/29/2022]
Abstract
Reverse genetics in Leishmania spp has gained importance beyond basic research as efforts increase to discover and validate new drug targets. Often, the most promising targets are essential for viability of the parasites, defying a genetic analysis by current gene replacement strategies. Duncan et al. demonstrate the applicability of DiCre recombination in Leishmania for induced replacement of the kinase CRK3 gene in promastigotes. DiCre gene replacement leads to the rapid loss of the gene and allows monitoring the phenotypic effects of the loss of function, eliminating the need for prolonged cultivation and selection. Implementation of the DiCre approach will allow functional genetics of the most important of Leishmania genes and is likely to boost genetic research and drug target discovery.
Collapse
Affiliation(s)
- Gerald F Späth
- Institut Pasteur and INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Paris, France
| | - Joachim Clos
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| |
Collapse
|
115
|
Duncan SM, Myburgh E, Philipon C, Brown E, Meissner M, Brewer J, Mottram JC. Conditional gene deletion with DiCre demonstrates an essential role for CRK3 in Leishmania mexicana cell cycle regulation. Mol Microbiol 2016; 100:931-44. [PMID: 26991545 PMCID: PMC4913733 DOI: 10.1111/mmi.13375] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2016] [Indexed: 12/22/2022]
Abstract
Leishmania mexicana has a large family of cyclin‐dependent kinases (CDKs) that reflect the complex interplay between cell cycle and life cycle progression. Evidence from previous studies indicated that Cdc2‐related kinase 3 (CRK3) in complex with the cyclin CYC6 is a functional homologue of the major cell cycle regulator CDK1, yet definitive genetic evidence for an essential role in parasite proliferation is lacking. To address this, we have implemented an inducible gene deletion system based on a dimerised Cre recombinase (diCre) to target CRK3 and elucidate its role in the cell cycle of L. mexicana. Induction of diCre activity in promastigotes with rapamycin resulted in efficient deletion of floxed CRK3, resulting in G2/M growth arrest. Co‐expression of a CRK3 transgene during rapamycin‐induced deletion of CRK3 resulted in complementation of growth, whereas expression of an active site CRK3T178E mutant did not, showing that protein kinase activity is crucial for CRK3 function. Inducible deletion of CRK3 in stationary phase promastigotes resulted in attenuated growth in mice, thereby confirming CRK3 as a useful therapeutic target and diCre as a valuable new tool for analyzing essential genes in Leishmania.
Collapse
Affiliation(s)
- Samuel M Duncan
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Elmarie Myburgh
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK.,Centre for Immunology and Infection, Department of Biology, University of York, Wentworth Way, Heslington, York, YO10 5DD, UK
| | - Cintia Philipon
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Elaine Brown
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK.,Centre for Immunology and Infection, Department of Biology, University of York, Wentworth Way, Heslington, York, YO10 5DD, UK
| | - Markus Meissner
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - James Brewer
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Jeremy C Mottram
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK.,Centre for Immunology and Infection, Department of Biology, University of York, Wentworth Way, Heslington, York, YO10 5DD, UK
| |
Collapse
|
116
|
Jones ML, Das S, Belda H, Collins CR, Blackman MJ, Treeck M. A versatile strategy for rapid conditional genome engineering using loxP sites in a small synthetic intron in Plasmodium falciparum. Sci Rep 2016; 6:21800. [PMID: 26892670 PMCID: PMC4759600 DOI: 10.1038/srep21800] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 02/01/2016] [Indexed: 11/09/2022] Open
|
117
|
Brochet M, Billker O. Calcium signalling in malaria parasites. Mol Microbiol 2016; 100:397-408. [PMID: 26748879 DOI: 10.1111/mmi.13324] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2016] [Indexed: 12/24/2022]
Abstract
Ca(2+) is a ubiquitous intracellular messenger in malaria parasites with important functions in asexual blood stages responsible for malaria symptoms, the preceding liver-stage infection and transmission through the mosquito. Intracellular messengers amplify signals by binding to effector molecules that trigger physiological changes. The characterisation of some Ca(2+) effector proteins has begun to provide insights into the vast range of biological processes controlled by Ca(2+) signalling in malaria parasites, including host cell egress and invasion, protein secretion, motility and cell cycle regulation. Despite the importance of Ca(2+) signalling during the life cycle of malaria parasites, little is known about Ca(2+) homeostasis. Recent findings highlighted that upstream of stage-specific Ca(2+) effectors is a conserved interplay between second messengers to control critical intracellular Ca(2+) signals throughout the life cycle. The identification of the molecular mechanisms integrating stage-transcending mechanisms of Ca(2+) homeostasis in a network of stage-specific regulator and effector pathways now represents a major challenge for a meaningful understanding of Ca(2+) signalling in malaria parasites.
Collapse
Affiliation(s)
- Mathieu Brochet
- Faculty of Medicine, Department of Microbiology and Molecular Medicine, University of Geneva, 1 Rue Michel-Servet, CH-1211 Geneva 4, Switzerland.,UMR5235 CNRS-Université Montpellier 2, 34095, Montpellier, France
| | - Oliver Billker
- Wellcome Trust Sanger Institute, Malaria Programme, CB10 1SA, Hinxton, UK
| |
Collapse
|
118
|
Abstract
Protein post-translational modifications (PTM) are commonly used to regulate biological processes. Protein S-acylation is an enzymatically regulated reversible modification that has been shown to modulate protein localization, activity and membrane binding. Proteome-scale discovery on Plasmodium falciparum schizonts has revealed a complement of more than 400 palmitoylated proteins, including those essential for host invasion and drug resistance. The wide regulatory affect on this species is endorsed by the presence of 12 proteins containing the conserved DHHC-CRD (DHHC motif within a cysteine-rich domain) that is associated with palmitoyl-transferase activity. Genetic interrogation of these enzymes in Apicomplexa has revealed essentiality and distinct localization at cellular compartments; these features are species specific and are not observed in yeast. It is clear that palmitoylation has an elaborate role in Plasmodium biology and opens intriguing questions on the functional consequence of this group of acylation modifications and how the protein S-acyl transferases (PATs) orchestrate molecular events.
Collapse
|
119
|
Riglar DT, Whitehead L, Cowman AF, Rogers KL, Baum J. Localisation-based imaging of malarial antigens during erythrocyte entry reaffirms a role for AMA1 but not MTRAP in invasion. J Cell Sci 2015; 129:228-42. [PMID: 26604223 PMCID: PMC4732298 DOI: 10.1242/jcs.177741] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 11/16/2015] [Indexed: 01/17/2023] Open
Abstract
Microscopy-based localisation of proteins during malaria parasite (Plasmodium) invasion of the erythrocyte is widely used for tentative assignment of protein function. To date, however, imaging has been limited by the rarity of invasion events and the poor resolution available, given the micron size of the parasite, which leads to a lack of quantitative measures for definitive localisation. Here, using computational image analysis we have attempted to assign relative protein localisation during invasion using wide-field deconvolution microscopy. By incorporating three-dimensional information we present a detailed assessment of known parasite effectors predicted to function during entry but as yet untested or for which data are equivocal. Our method, termed longitudinal intensity profiling, resolves confusion surrounding the localisation of apical membrane antigen 1 (AMA1) at the merozoite–erythrocyte junction and predicts that the merozoite thrombospondin-related anonymous protein (MTRAP) is unlikely to play a direct role in the mechanics of entry, an observation supported with additional biochemical evidence. This approach sets a benchmark for imaging of complex micron-scale events and cautions against simplistic interpretations of small numbers of representative images for the assignment of protein function or prioritisation of candidates as therapeutic targets. Highlighted Article: Here we develop a high-definition imaging approach to dissect and assign function to proteins involved in the rapid process of malaria parasite invasion of the human erythrocyte.
Collapse
Affiliation(s)
- David T Riglar
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, 3052, Australia Department of Medical Biology, University of Melbourne, Victoria, 3050, Melbourne, Australia
| | - Lachlan Whitehead
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, 3052, Australia Department of Medical Biology, University of Melbourne, Victoria, 3050, Melbourne, Australia
| | - Alan F Cowman
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, 3052, Australia Department of Medical Biology, University of Melbourne, Victoria, 3050, Melbourne, Australia
| | - Kelly L Rogers
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, 3052, Australia Department of Medical Biology, University of Melbourne, Victoria, 3050, Melbourne, Australia
| | - Jake Baum
- Department of Life Sciences, Imperial College, London SW7 2AZ, UK
| |
Collapse
|
120
|
Hallée S, Richard D. Evidence that the Malaria Parasite Plasmodium falciparum Putative Rhoptry Protein 2 Localizes to the Golgi Apparatus throughout the Erythrocytic Cycle. PLoS One 2015; 10:e0138626. [PMID: 26375591 PMCID: PMC4574476 DOI: 10.1371/journal.pone.0138626] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 08/31/2015] [Indexed: 11/28/2022] Open
Abstract
Invasion of a red blood cell by Plasmodium falciparum merozoites is an essential step in the malaria lifecycle. Several of the proteins involved in this process are stored in the apical complex of the merozoite, a structure containing secretory organelles that are released at specific times during invasion. The molecular players involved in erythrocyte invasion thus represent potential key targets for both therapeutic and vaccine-based strategies to block parasite development. In our quest to identify and characterize new effectors of invasion, we investigated the P. falciparum homologue of a P. berghei protein putatively localized to the rhoptries, the Putative rhoptry protein 2 (PbPRP2). We show that in P. falciparum, the protein colocalizes extensively with the Golgi apparatus across the asexual erythrocytic cycle. Furthermore, imaging of merozoites caught at different times during invasion show that PfPRP2 is not secreted during the process instead staying associated with the Golgi apparatus. Our evidence therefore suggests that PfPRP2 is a Golgi protein and that it is likely not a direct effector in the process of merozoite invasion.
Collapse
Affiliation(s)
- Stéphanie Hallée
- Centre de recherche en infectiologie, CHU-Université Laval, Quebec City, Quebec, Canada
| | - Dave Richard
- Centre de recherche en infectiologie, CHU-Université Laval, Quebec City, Quebec, Canada
- * E-mail:
| |
Collapse
|
121
|
Mitcheson DF, Tobin AB, Alam MM. Applying chemical genetic tools to the study of phospho-signalling pathways in malaria parasites. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:1650-6. [PMID: 26143498 DOI: 10.1016/j.bbapap.2015.06.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 06/25/2015] [Accepted: 06/30/2015] [Indexed: 12/27/2022]
Abstract
Until very recently there has been very little information about the phospho-signalling pathways in apicomplexan parasites including the most virulent species of human malaria parasite, Plasmodium falciparum. With the advancement of mass spectrometry-based phosphoproteomics and the development of chemical genetic approaches to target specific parasite protein kinases, the complexity of the essential role played by phosphorylation in maintaining the viability of apicomplexan parasites is now being revealed. This review will describe these recent advances and will discuss how these approaches can be used to validate parasite protein kinases as drug targets and to determine the on- and off-target action of protein kinase inhibitors. This article is part of a Special Issue entitled: Inhibitors of Protein Kinases.
Collapse
Affiliation(s)
- Deborah F Mitcheson
- Department of Cell Physiology and Pharmacology, University of Leicester, Hodgkin Building, Lancaster Road, Leicester LE1 9HN, UK
| | - Andrew B Tobin
- MRC Toxicology Unit, University of Leicester, Hodgkin Building, Lancaster Road, Leicester LE1 9HN, UK
| | - Mahmood M Alam
- MRC Toxicology Unit, University of Leicester, Hodgkin Building, Lancaster Road, Leicester LE1 9HN, UK.
| |
Collapse
|
122
|
Philip N, Waters AP. Conditional Degradation of Plasmodium Calcineurin Reveals Functions in Parasite Colonization of both Host and Vector. Cell Host Microbe 2015; 18:122-31. [PMID: 26118994 PMCID: PMC4509507 DOI: 10.1016/j.chom.2015.05.018] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 04/29/2015] [Accepted: 05/27/2015] [Indexed: 12/01/2022]
Abstract
Functional analysis of essential genes in the malarial parasite, Plasmodium, is hindered by lack of efficient strategies for conditional protein regulation. We report the development of a rapid, specific, and inducible chemical-genetic tool in the rodent malaria parasite, P. berghei, in which endogenous proteins engineered to contain the auxin-inducible degron (AID) are selectively degraded upon adding auxin. Application of AID to the calcium-regulated protein phosphatase, calcineurin, revealed functions in host and vector stages of parasite development. Whereas depletion of calcineurin in late-stage schizonts demonstrated its critical role in erythrocyte attachment and invasion in vivo, stage-specific depletion uncovered roles in gamete development, fertilization, and ookinete-to-oocyst and sporozoite-to-liver stage transitions. Furthermore, AID technology facilitated concurrent generation and phenotyping of transgenic lines, allowing multiple lines to be assessed simultaneously with significant reductions in animal use. This study highlights the broad applicability of AID for functional analysis of proteins across the Plasmodium life cycle. Calcineurin regulates colonization of host cells across the Plasmodium life cycle Calcineurin regulates male gametogenesis AID technology is broadly applicable to study protein function in Plasmodium Multiplexing of AID technology results in substantially reduced animal use
Collapse
Affiliation(s)
- Nisha Philip
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical Veterinary & Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK.
| | - Andrew P Waters
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical Veterinary & Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK.
| |
Collapse
|
123
|
Pieperhoff MS, Pall GS, Jiménez-Ruiz E, Das S, Melatti C, Gow M, Wong EH, Heng J, Müller S, Blackman MJ, Meissner M. Conditional U1 Gene Silencing in Toxoplasma gondii. PLoS One 2015; 10:e0130356. [PMID: 26090798 PMCID: PMC4474610 DOI: 10.1371/journal.pone.0130356] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 05/18/2015] [Indexed: 11/19/2022] Open
Abstract
The functional characterisation of essential genes in apicomplexan parasites, such as Toxoplasma gondii or Plasmodium falciparum, relies on conditional mutagenesis systems. Here we present a novel strategy based on U1 snRNP-mediated gene silencing. U1 snRNP is critical in pre-mRNA splicing by defining the exon-intron boundaries. When a U1 recognition site is placed into the 3'-terminal exon or adjacent to the termination codon, pre-mRNA is cleaved at the 3'-end and degraded, leading to an efficient knockdown of the gene of interest (GOI). Here we describe a simple method that combines endogenous tagging with DiCre-mediated positioning of U1 recognition sites adjacent to the termination codon of the GOI which leads to a conditional knockdown of the GOI upon rapamycin-induction. Specific knockdown mutants of the reporter gene GFP and several endogenous genes of T. gondii including the clathrin heavy chain gene 1 (chc1), the vacuolar protein sorting gene 26 (vps26), and the dynamin-related protein C gene (drpC) were silenced using this approach and demonstrate the potential of this technology. We also discuss advantages and disadvantages of this method in comparison to other technologies in more detail.
Collapse
Affiliation(s)
- Manuela S. Pieperhoff
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow, Lanarkshire, United Kingdom
| | - Gurman S. Pall
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow, Lanarkshire, United Kingdom
| | - Elena Jiménez-Ruiz
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow, Lanarkshire, United Kingdom
| | - Sujaan Das
- Division of Parasitology, MRC National Institute for Medical Research, Mill Hill, London, United Kingdom
| | - Carmen Melatti
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow, Lanarkshire, United Kingdom
| | - Matthew Gow
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow, Lanarkshire, United Kingdom
| | - Eleanor H. Wong
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow, Lanarkshire, United Kingdom
| | - Joanne Heng
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow, Lanarkshire, United Kingdom
| | - Sylke Müller
- Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow, Lanarkshire, United Kingdom
| | - Michael J. Blackman
- Division of Parasitology, MRC National Institute for Medical Research, Mill Hill, London, United Kingdom
- * E-mail: (MM); (MJB)
| | - Markus Meissner
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow, Lanarkshire, United Kingdom
- * E-mail: (MM); (MJB)
| |
Collapse
|
124
|
Role and Regulation of Glutathione Metabolism in Plasmodium falciparum. Molecules 2015; 20:10511-34. [PMID: 26060916 PMCID: PMC6272303 DOI: 10.3390/molecules200610511] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 05/11/2015] [Accepted: 06/01/2015] [Indexed: 11/30/2022] Open
Abstract
Malaria in humans is caused by one of five species of obligate intracellular protozoan parasites of the genus Plasmodium. P. falciparum causes the most severe disease and is responsible for 600,000 deaths annually, primarily in Sub-Saharan Africa. It has long been suggested that during their development, malaria parasites are exposed to environmental and metabolic stresses. One strategy to drug discovery was to increase these stresses by interfering with the parasites’ antioxidant and redox systems, which may be a valuable approach to disease intervention. Plasmodium possesses two redox systems—the thioredoxin and the glutathione system—with overlapping but also distinct functions. Glutathione is the most abundant low molecular weight redox active thiol in the parasites existing primarily in its reduced form representing an excellent thiol redox buffer. This allows for an efficient maintenance of the intracellular reducing environment of the parasite cytoplasm and its organelles. This review will highlight the mechanisms that are responsible for sustaining an adequate concentration of glutathione and maintaining its redox state in Plasmodium. It will provide a summary of the functions of the tripeptide and will discuss the potential of glutathione metabolism for drug discovery against human malaria parasites.
Collapse
|
125
|
de Koning-Ward TF, Gilson PR, Crabb BS. Advances in molecular genetic systems in malaria. Nat Rev Microbiol 2015; 13:373-87. [DOI: 10.1038/nrmicro3450] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
126
|
Stallmach R, Kavishwar M, Withers-Martinez C, Hackett F, Collins CR, Howell SA, Yeoh S, Knuepfer E, Atid AJ, Holder AA, Blackman MJ. Plasmodium falciparum SERA5 plays a non-enzymatic role in the malarial asexual blood-stage lifecycle. Mol Microbiol 2015; 96:368-87. [PMID: 25599609 PMCID: PMC4671257 DOI: 10.1111/mmi.12941] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2015] [Indexed: 02/02/2023]
Abstract
The malaria parasite Plasmodium falciparum replicates in an intraerythrocytic parasitophorous vacuole (PV). The most abundant P. falciparum PV protein, called SERA5, is essential in blood stages and possesses a papain-like domain, prompting speculation that it functions as a proteolytic enzyme. Unusually however, SERA5 possesses a Ser residue (Ser596) at the position of the canonical catalytic Cys of papain-like proteases, and the function of SERA5 or whether it performs an enzymatic role is unknown. In this study, we failed to detect proteolytic activity associated with the Ser596-containing parasite-derived or recombinant protein. However, substitution of Ser596 with a Cys residue produced an active recombinant enzyme with characteristics of a cysteine protease, demonstrating that SERA5 can bind peptides. Using targeted homologous recombination in P. falciparum, we substituted Ser596 with Ala with no phenotypic consequences, proving that SERA5 does not perform an essential enzymatic role in the parasite. We could also replace an internal segment of SERA5 with an affinity-purification tag. In contrast, using almost identical targeting constructs, we could not truncate or C-terminally tag the SERA5 gene, or replace Ser596 with a bulky Arg residue. Our findings show that SERA5 plays an indispensable but non-enzymatic role in the P. falciparum blood-stage life cycle.
Collapse
Affiliation(s)
- Robert Stallmach
- Division of Parasitology, MRC National Institute for Medical ResearchLondon, NW7 1AA, UK
| | - Manoli Kavishwar
- Division of Parasitology, MRC National Institute for Medical ResearchLondon, NW7 1AA, UK
| | | | - Fiona Hackett
- Division of Parasitology, MRC National Institute for Medical ResearchLondon, NW7 1AA, UK
| | - Christine R Collins
- Division of Parasitology, MRC National Institute for Medical ResearchLondon, NW7 1AA, UK
| | - Steven A Howell
- Division of Molecular Structure, MRC National Institute for Medical ResearchLondon, NW7 1AA, UK
| | - Sharon Yeoh
- Division of Parasitology, MRC National Institute for Medical ResearchLondon, NW7 1AA, UK
| | - Ellen Knuepfer
- Division of Parasitology, MRC National Institute for Medical ResearchLondon, NW7 1AA, UK
| | - Avshalom J Atid
- Division of Parasitology, MRC National Institute for Medical ResearchLondon, NW7 1AA, UK
| | - Anthony A Holder
- Division of Parasitology, MRC National Institute for Medical ResearchLondon, NW7 1AA, UK
| | - Michael J Blackman
- Division of Parasitology, MRC National Institute for Medical ResearchLondon, NW7 1AA, UK
| |
Collapse
|
127
|
Gomes AR, Bushell E, Schwach F, Girling G, Anar B, Quail MA, Herd C, Pfander C, Modrzynska K, Rayner JC, Billker O. A genome-scale vector resource enables high-throughput reverse genetic screening in a malaria parasite. Cell Host Microbe 2015; 17:404-413. [PMID: 25732065 PMCID: PMC4362957 DOI: 10.1016/j.chom.2015.01.014] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 12/01/2014] [Accepted: 01/07/2015] [Indexed: 10/30/2022]
Abstract
The genome-wide identification of gene functions in malaria parasites is hampered by a lack of reverse genetic screening methods. We present a large-scale resource of barcoded vectors with long homology arms for effective modification of the Plasmodium berghei genome. Cotransfecting dozens of vectors into the haploid blood stages creates complex pools of barcoded mutants, whose competitive fitness can be measured during infection of a single mouse using barcode sequencing (barseq). To validate the utility of this resource, we rescreen the P. berghei kinome, using published kinome screens for comparison. We find that several protein kinases function redundantly in asexual blood stages and confirm the targetability of kinases cdpk1, gsk3, tkl3, and PBANKA_082960 by genotyping cloned mutants. Thus, parallel phenotyping of barcoded mutants unlocks the power of reverse genetic screening for a malaria parasite and will enable the systematic identification of genes essential for in vivo parasite growth and transmission.
Collapse
Affiliation(s)
- Ana Rita Gomes
- Wellcome Trust Sanger Institute, Hinxton Cambridge CB10 1SA, UK
| | - Ellen Bushell
- Wellcome Trust Sanger Institute, Hinxton Cambridge CB10 1SA, UK
| | - Frank Schwach
- Wellcome Trust Sanger Institute, Hinxton Cambridge CB10 1SA, UK
| | - Gareth Girling
- Wellcome Trust Sanger Institute, Hinxton Cambridge CB10 1SA, UK
| | - Burcu Anar
- Wellcome Trust Sanger Institute, Hinxton Cambridge CB10 1SA, UK
| | - Michael A Quail
- Wellcome Trust Sanger Institute, Hinxton Cambridge CB10 1SA, UK
| | - Colin Herd
- Wellcome Trust Sanger Institute, Hinxton Cambridge CB10 1SA, UK
| | - Claudia Pfander
- Wellcome Trust Sanger Institute, Hinxton Cambridge CB10 1SA, UK
| | | | - Julian C Rayner
- Wellcome Trust Sanger Institute, Hinxton Cambridge CB10 1SA, UK.
| | - Oliver Billker
- Wellcome Trust Sanger Institute, Hinxton Cambridge CB10 1SA, UK.
| |
Collapse
|
128
|
Conway DJ. Paths to a malaria vaccine illuminated by parasite genomics. Trends Genet 2015; 31:97-107. [PMID: 25620796 PMCID: PMC4359294 DOI: 10.1016/j.tig.2014.12.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 12/19/2014] [Accepted: 12/19/2014] [Indexed: 11/24/2022]
Abstract
Discovery of vaccine candidate antigens by parasite genome sequence analyses. Genetic crosses, linkage group selection, and functional studies on parasites. Characterizing developmental and epigenetic variation alongside allelic polymorphism. Selection by naturally acquired immune responses helps to focus vaccine design.
More human death and disease is caused by malaria parasites than by all other eukaryotic pathogens combined. As early as the sequencing of the first human genome, malaria parasite genomics was prioritized to fuel the discovery of vaccine candidate antigens. This stimulated increased research on malaria, generating new understanding of the cellular and molecular mechanisms of infection and immunity. This review of recent developments illustrates how new approaches in parasite genomics, and increasingly large amounts of data from population studies, are helping to identify antigens that are promising lead targets. Although these results have been encouraging, effective discovery and characterization need to be coupled with more innovation and funding to translate findings into newly designed vaccine products for clinical trials.
Collapse
Affiliation(s)
- David J Conway
- Pathogen Molecular Biology Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK.
| |
Collapse
|
129
|
Kangussu-Marcolino MM, Cunha AP, Avila AR, Herman JP, DaRocha WD. Conditional removal of selectable markers in Trypanosoma cruzi using a site-specific recombination tool: proof of concept. Mol Biochem Parasitol 2015; 198:71-4. [PMID: 25619800 DOI: 10.1016/j.molbiopara.2015.01.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 01/12/2015] [Accepted: 01/14/2015] [Indexed: 11/29/2022]
Abstract
The scarcity of molecular tools for genetic manipulation is a critical obstacle for functional genomics studies on Trypanosoma cruzi. The current study adapted an inducible site-specific recombination system based on Dimerizable CRE recombinase (DiCRE). Two vectors for stable transfection were created, a first one to express inactive portions of DiCRE recombinase, and a second plasmid containing the loxP sites to test DiCRE activity. After integrating both constructs into the T. cruzi genome, it was shown that DiCRE recombinase can be efficiently used to manipulate its genome by allowing the removal of selectable markers thus generating homogeneous populations. The DiCRE recombinase success allows conditional knockout and the removal of selectable markers without prior parasite modification, which also facilitate the transferring of DiCRE recombinase to different T. cruzi strains.
Collapse
Affiliation(s)
| | - Ana Paula Cunha
- Departamento de Bioquímica e Biologia Molecular, UFPR, Brazil
| | | | | | | |
Collapse
|
130
|
Goldfless SJ, Wagner JC, Niles JC. Versatile control of Plasmodium falciparum gene expression with an inducible protein-RNA interaction. Nat Commun 2014; 5:5329. [PMID: 25370483 PMCID: PMC4223869 DOI: 10.1038/ncomms6329] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 09/20/2014] [Indexed: 01/06/2023] Open
Abstract
The available tools for conditional gene expression in Plasmodium falciparum are limited. Here, to enable reliable control of target gene expression, we build a system to efficiently modulate translation. We overcame several problems associated with other approaches for regulating gene expression in P. falciparum. Specifically, our system functions predictably across several native and engineered promoter contexts, and affords control over reporter and native parasite proteins irrespective of their subcellular compartmentalization. Induction and repression of gene expression are rapid, homogeneous and stable over prolonged periods. To demonstrate practical application of our system, we used it to reveal direct links between antimalarial drugs and their native parasite molecular target. This is an important outcome given the rapid spread of resistance, and intensified efforts to efficiently discover and optimize new antimalarial drugs. Overall, the studies presented highlight the utility of our system for broadly controlling gene expression and performing functional genetics in P. falciparum.
Collapse
Affiliation(s)
- Stephen J Goldfless
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
| | - Jeffrey C Wagner
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
| | - Jacquin C Niles
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
131
|
Webster WAJ, McFadden GI. From the genome to the phenome: tools to understand the basic biology of Plasmodium falciparum. J Eukaryot Microbiol 2014; 61:655-71. [PMID: 25227912 DOI: 10.1111/jeu.12176] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 09/01/2014] [Accepted: 09/02/2014] [Indexed: 11/30/2022]
Abstract
Malaria plagues one out of every 30 humans and contributes to almost a million deaths, and the problem could worsen. Our current therapeutic options are compromised by emerging resistance by the parasite to our front line drugs. It is thus imperative to better understand the basic biology of the parasite and develop novel drugs to stem this disease. The most facile approach to analyse a gene's function is to remove it from the genome or inhibit its activity. Although genetic manipulation of the human malaria parasite Plasmodium falciparum is a relatively standard procedure, there is no optimal method to perturb genes essential to the intraerythrocytic development cycle--the part of the life cycle that produces the clinical manifestation of malaria. This is a severe impediment to progress because the phenotype we wish to study is exactly the one that is so elusive. In the absence of any utilitarian way to conditionally delete essential genes, we are prevented from investigating the parasite's most vulnerable points. This review aims to focus on the development of tools identifying essential genes of P. falciparum and our ability to elicit phenotypic mutation.
Collapse
Affiliation(s)
- Wesley A J Webster
- Centre for Regional and Rural Futures, School of Life and Environmental Sciences, Deakin University, Burwood, 3125, Victoria, Australia; Plant Cell Biology Research Centre, School of Botany, University of Melbourne, Melbourne, 3010, Victoria, Australia
| | | |
Collapse
|
132
|
Sudarsan R, Chopra RK, Khan MA, Sharma S. Ribosomal protein P2 localizes to the parasite zoite-surface and is a target for invasion inhibitory antibodies in Toxoplasma gondii and Plasmodium falciparum. Parasitol Int 2014; 64:43-9. [PMID: 25280460 DOI: 10.1016/j.parint.2014.08.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 08/14/2014] [Accepted: 08/30/2014] [Indexed: 10/24/2022]
Abstract
In the malarial parasite Plasmodium falciparum, the conserved ribosomal stalk protein P2 (PfP2) exhibits extra-ribosomal stage-specific oligomerization and trafficking to the host red cell membrane. Antibodies directed against PfP2 arrested cell division. We sought to examine whether P2 from a closely related Apicomplexan parasite, Toxoplasma gondii, exhibits similar properties in terms of its oligomeric status as well as such unique host-cell localization. Circular dichroism spectroscopy of recombinant P2 from T. gondii (TgP2) showed a structure similar to that of PfP2, but unlike PfP2, which forms SDS- and DTT-resistant oligomers, TgP2 exhibited only a weak SDS-resistant dimerization. Also, unlike PfP2 localization to the infected erythrocyte surface, TgP2 did not localize to the host membrane in T. gondii infected human foreskin fibroblast cells. However, P2 protein was detected on the free tachyzoite surface, corroborated by localization of epitope-tagged P2 transfected in T. gondii. The presence of P2 on the surface of P. falciparum merozoites was also observed, and specific antibodies raised against the P2 protein blocked both T. gondii and P. falciparum zoite invasion of the host cells. Thus, although certain moonlighting functions of the acidic ribosomal protein P2 are different amongst P. falciparum and T. gondii, the P2 protein localizes to the surface of the invasive zoite form, and appears to constitute a potential target for host cell invasion inhibition in both the Apicomplexan infections.
Collapse
Affiliation(s)
- Rajagopal Sudarsan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India.
| | - Reshma Korde Chopra
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Mudassar Ali Khan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Shobhona Sharma
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
133
|
Advantages and disadvantages of conditional systems for characterization of essential genes in Toxoplasma gondii. Parasitology 2014; 141:1390-8. [PMID: 24926834 DOI: 10.1017/s0031182014000559] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The dissection of apicomplexan biology has been highly influenced by the genetic tools available for manipulation of parasite DNA. Here, we describe different techniques available for the generation of conditional mutants. Comparison of the advantages and disadvantages of the three most commonly used regulation systems: the tetracycline inducible system, the regulation of protein stability and site-specific recombination are discussed. Using some previously described examples we explore some of the pitfalls involved in gene-function analysis using these systems that can lead to wrong or over-interpretation of phenotypes. We will also mention different options to standardize the application of these techniques for the characterization of gene function in high-throughput.
Collapse
|
134
|
Genome editing in the human malaria parasite Plasmodium falciparum using the CRISPR-Cas9 system. Nat Biotechnol 2014; 32:819-21. [DOI: 10.1038/nbt.2925] [Citation(s) in RCA: 474] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 05/08/2014] [Indexed: 02/06/2023]
|
135
|
Yap A, Azevedo MF, Gilson PR, Weiss GE, O'Neill MT, Wilson DW, Crabb BS, Cowman AF. Conditional expression of apical membrane antigen 1 in Plasmodium falciparum shows it is required for erythrocyte invasion by merozoites. Cell Microbiol 2014; 16:642-56. [PMID: 24571085 PMCID: PMC4231980 DOI: 10.1111/cmi.12287] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 02/23/2014] [Accepted: 02/24/2014] [Indexed: 11/30/2022]
Abstract
Malaria is caused by obligate intracellular parasites, of which Plasmodium falciparum is the most lethal species. In humans, P. falciparum merozoites (invasive forms of the parasite) employ a host of parasite proteins to rapidly invade erythrocytes. One of these is the P. falciparum apical membrane antigen 1 (PfAMA1) which forms a complex with rhoptry neck proteins at the tight junction. Here, we have placed the Pfama1 gene under conditional control using dimerizable Cre recombinase (DiCre) in P. falciparum. DiCre-mediated excision of the loxP-flanked Pfama1 gene results in approximately 80% decreased expression of the protein within one intraerythrocytic growth cycle. This reduces growth by 40%, due to decreased invasion efficiency characterized by a post-invasion defect in sealing of the parasitophorous vacuole. These results show that PfAMA1 is an essential protein for merozoite invasion in P. falciparum and either directly or indirectly plays a role in resealing of the red blood cell at the posterior end of the invasion event.
Collapse
Affiliation(s)
- Alan Yap
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Vic., 3052, Australia
| | | | | | | | | | | | | | | |
Collapse
|
136
|
Honma H, Hirai M, Nakamura S, Hakimi H, Kawazu SI, Palacpac NMQ, Hisaeda H, Matsuoka H, Kawai S, Endo H, Yasunaga T, Ohashi J, Mita T, Horii T, Furusawa M, Tanabe K. Generation of rodent malaria parasites with a high mutation rate by destructing proofreading activity of DNA polymerase δ. DNA Res 2014; 21:439-46. [PMID: 24670267 PMCID: PMC4131837 DOI: 10.1093/dnares/dsu009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Plasmodium falciparum malaria imposes a serious public health concern throughout the tropics. Although genetic tools are principally important to fully investigate malaria parasites, currently available forward and reverse tools are fairly limited. It is expected that parasites with a high mutation rate can readily acquire novel phenotypes/traits; however, they remain an untapped tool for malaria biology. Here, we generated a mutator malaria parasite (hereinafter called a ‘malaria mutator’), using site-directed mutagenesis and gene transfection techniques. A mutator Plasmodium berghei line with a defective proofreading 3′ → 5′ exonuclease activity in DNA polymerase δ (referred to as PbMut) and a control P. berghei line with wild-type DNA polymerase δ (referred to as PbCtl) were maintained by weekly passage in ddY mice for 122 weeks. High-throughput genome sequencing analysis revealed that two PbMut lines had 175–178 mutations and a 86- to 90-fold higher mutation rate than that of a PbCtl line. PbMut, PbCtl, and their parent strain, PbWT, showed similar course of infection. Interestingly, PbMut lost the ability to form gametocytes during serial passages. We believe that the malaria mutator system could provide a novel and useful tool to investigate malaria biology.
Collapse
Affiliation(s)
- Hajime Honma
- Laboratory of Malariology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Makoto Hirai
- Department of Parasitology, Graduate School of Medicine, Gunma University, Maebashi, Gunma 371-8511, Japan
| | - Shota Nakamura
- Department of Genome Informatics, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hassan Hakimi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada, Obihiro, Hokkaido 080-8555, Japan
| | - Shin-Ichiro Kawazu
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada, Obihiro, Hokkaido 080-8555, Japan
| | - Nirianne M Q Palacpac
- Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hajime Hisaeda
- Department of Parasitology, Graduate School of Medicine, Gunma University, Maebashi, Gunma 371-8511, Japan
| | - Hiroyuki Matsuoka
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Satoru Kawai
- Laboratory of Tropical Medicine and Parasitology, Institute of International Education and Research, Dokkyo Medical University, Shimotsuga, Tochigi 321-0293, Japan
| | - Hiroyoshi Endo
- Department of International Affairs and Tropical Medicine, Tokyo Women's Medical University School of Medicine, Kawada-cho, Shinjuku, Tokyo 162-8666, Japan
| | - Teruo Yasunaga
- Department of Genome Informatics, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Jun Ohashi
- Faculty of Medicine, University of Tsukuba, Ibaragi 305-8575, Japan
| | - Toshihiro Mita
- Department of International Affairs and Tropical Medicine, Tokyo Women's Medical University School of Medicine, Kawada-cho, Shinjuku, Tokyo 162-8666, Japan Department of Molecular and Cellular Parasitology, Juntendo University School of Medicine, Hongo, Bunkyo, Tokyo 113-8421, Japan
| | - Toshihiro Horii
- Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Mitsuru Furusawa
- Neo-Morgan Laboratory, Inc., Nogawa, Miyamae, Kawasaki, Kanagawa 216-0001, Japan
| | - Kazuyuki Tanabe
- Laboratory of Malariology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
137
|
Grüring C, Moon RW, Lim C, Holder AA, Blackman MJ, Duraisingh MT. Human red blood cell-adapted Plasmodium knowlesi parasites: a new model system for malaria research. Cell Microbiol 2014; 16:612-20. [PMID: 24506567 DOI: 10.1111/cmi.12275] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/24/2014] [Accepted: 01/27/2014] [Indexed: 02/06/2023]
Abstract
Plasmodium knowlesi is a simian malaria parasite primarily infecting macaque species in Southeast Asia. Although its capacity to infect humans has been recognized since the early part of the last century, it has recently become evident that human infections are widespread and potentially life threatening. Historically, P. knowlesi has proven to be a powerful tool in early studies of malaria parasites, providing key breakthroughs in understanding many aspects of Plasmodium biology. However, the necessity to grow the parasite either in macaques or in vitro using macaque blood restricted research to laboratories with access to these resources. The recent adaptation of P. knowlesi to grow and proliferate in vitro in human red blood cells (RBCs) is therefore a substantial step towards revitalizing and expanding research on P. knowlesi. Furthermore, the development of a highly efficient transfection system to genetically modify the parasite makes P. knowlesi an ideal model to study parasite biology. In this review, we elaborate on the importance of P. knowlesi in earlier phases of malaria research and highlight the future potential of the newly available human adapted P. knowlesi parasite lines.
Collapse
Affiliation(s)
- Christof Grüring
- Department of Immunology & Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA
| | | | | | | | | | | |
Collapse
|
138
|
Abstract
Toxoplasma gondii and Plasmodium falciparum are important human pathogens. These parasites and many of their apicomplexan relatives undergo a complex developmental process in the cells of their hosts, which includes genome replication, cell division and the assembly of new invasive stages. Apicomplexan cell cycle progression is both globally and locally regulated. Global regulation is carried out throughout the cytoplasm by diffusible factors that include cell cycle-specific kinases, cyclins and transcription factors. Local regulation acts on individual nuclei and daughter cells that are developing inside the mother cell. We propose that the centrosome is a master regulator that physically tethers cellular components and that provides spatial and temporal control of apicomplexan cell division.
Collapse
Affiliation(s)
- Maria E Francia
- Department of Cellular Biology, University of Georgia, Athens, Georgia 30602, USA
| | - Boris Striepen
- 1] Department of Cellular Biology, University of Georgia, Athens, Georgia 30602, USA. [2] Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia 30602, USA
| |
Collapse
|
139
|
Suarez C, Volkmann K, Gomes AR, Billker O, Blackman MJ. The malarial serine protease SUB1 plays an essential role in parasite liver stage development. PLoS Pathog 2013; 9:e1003811. [PMID: 24348254 PMCID: PMC3861531 DOI: 10.1371/journal.ppat.1003811] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 10/21/2013] [Indexed: 11/30/2022] Open
Abstract
Transmission of the malaria parasite to its vertebrate host involves an obligatory exoerythrocytic stage in which extensive asexual replication of the parasite takes place in infected hepatocytes. The resulting liver schizont undergoes segmentation to produce thousands of daughter merozoites. These are released to initiate the blood stage life cycle, which causes all the pathology associated with the disease. Whilst elements of liver stage merozoite biology are similar to those in the much better-studied blood stage merozoites, little is known of the molecular players involved in liver stage merozoite production. To facilitate the study of liver stage biology we developed a strategy for the rapid production of complex conditional alleles by recombinase mediated engineering in Escherichia coli, which we used in combination with existing Plasmodium berghei deleter lines expressing Flp recombinase to study subtilisin-like protease 1 (SUB1), a conserved Plasmodium serine protease previously implicated in blood stage merozoite maturation and egress. We demonstrate that SUB1 is not required for the early stages of intrahepatic growth, but is essential for complete development of the liver stage schizont and for production of hepatic merozoites. Our results indicate that inhibitors of SUB1 could be used in prophylactic approaches to control or block the clinically silent pre-erythrocytic stage of the malaria parasite life cycle. Malaria is caused by a single-celled parasite and is transmitted by the bite of an infected mosquito. The inoculated sporozoite forms of the parasite invade liver cells where they replicate, eventually releasing thousands of merozoites into the bloodstream to initiate the blood stage parasite life cycle which causes clinical malaria. The liver stage of the parasite life cycle is asymptomatic, so it is widely considered a potential target for prophylactic vaccine- or drug-based approaches designed to prevent infection. In this study, we use a robust, highly efficient gene engineering approach called recombineering, combined with a conditional gene deletion strategy to examine the function in liver stages of a parasite protease called SUB1, previously implicated in release of blood stage parasites. We show that SUB1 is expressed in the liver stage schizont and that the protease is essential for production of liver stage merozoites. Our results enhance our understanding of malarial liver stage biology, provide new tools for studying essential gene function in malaria, and suggest that inhibitors of SUB1 could be used as prophylactic drugs to prevent clinical malaria.
Collapse
Affiliation(s)
- Catherine Suarez
- Division of Parasitology, Medical Research Council National Institute for Medical Research, Mill Hill, London, United Kingdom
| | - Katrin Volkmann
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - Ana Rita Gomes
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - Oliver Billker
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom
- * E-mail: (OB); (MJB)
| | - Michael J. Blackman
- Division of Parasitology, Medical Research Council National Institute for Medical Research, Mill Hill, London, United Kingdom
- * E-mail: (OB); (MJB)
| |
Collapse
|
140
|
Type II fatty acid biosynthesis is essential for Plasmodium falciparum sporozoite development in the midgut of Anopheles mosquitoes. EUKARYOTIC CELL 2013; 13:550-9. [PMID: 24297444 DOI: 10.1128/ec.00264-13] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The prodigious rate at which malaria parasites proliferate during asexual blood-stage replication, midgut sporozoite production, and intrahepatic development creates a substantial requirement for essential nutrients, including fatty acids that likely are necessary for parasite membrane formation. Plasmodium parasites obtain fatty acids either by scavenging from the vertebrate host and mosquito vector or by producing fatty acids de novo via the type two fatty acid biosynthesis pathway (FAS-II). Here, we study the FAS-II pathway in Plasmodium falciparum, the species responsible for the most lethal form of human malaria. Using antibodies, we find that the FAS-II enzyme FabI is expressed in mosquito midgut oocysts and sporozoites as well as liver-stage parasites but not during the blood stages. As expected, FabI colocalizes with the apicoplast-targeted acyl carrier protein, indicating that FabI functions in the apicoplast. We further analyze the FAS-II pathway in Plasmodium falciparum by assessing the functional consequences of deleting fabI and fabB/F. Targeted deletion or disruption of these genes in P. falciparum did not affect asexual blood-stage replication or the generation of midgut oocysts; however, subsequent sporozoite development was abolished. We conclude that the P. falciparum FAS-II pathway is essential for sporozoite development within the midgut oocyst. These findings reveal an important distinction from the rodent Plasmodium parasites P. berghei and P. yoelii, where the FAS-II pathway is known to be required for normal parasite progression through the liver stage but is not required for oocyst development in the Anopheles mosquito midgut.
Collapse
|
141
|
Poulin B, Patzewitz EM, Brady D, Silvie O, Wright MH, Ferguson DJP, Wall RJ, Whipple S, Guttery DS, Tate EW, Wickstead B, Holder AA, Tewari R. Unique apicomplexan IMC sub-compartment proteins are early markers for apical polarity in the malaria parasite. Biol Open 2013; 2:1160-70. [PMID: 24244852 PMCID: PMC3828762 DOI: 10.1242/bio.20136163] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 08/21/2013] [Indexed: 11/20/2022] Open
Abstract
The phylum Apicomplexa comprises over 5000 intracellular protozoan parasites, including Plasmodium and Toxoplasma, that are clinically important pathogens affecting humans and livestock. Malaria parasites belonging to the genus Plasmodium possess a pellicle comprised of a plasmalemma and inner membrane complex (IMC), which is implicated in parasite motility and invasion. Using live cell imaging and reverse genetics in the rodent malaria model P. berghei, we localise two unique IMC sub-compartment proteins (ISPs) and examine their role in defining apical polarity during zygote (ookinete) development. We show that these proteins localise to the anterior apical end of the parasite where IMC organisation is initiated, and are expressed at all developmental stages, especially those that are invasive. Both ISP proteins are N-myristoylated, phosphorylated and membrane-bound. Gene disruption studies suggest that ISP1 is likely essential for parasite development, whereas ISP3 is not. However, an absence of ISP3 alters the apical localisation of ISP1 in all invasive stages including ookinetes and sporozoites, suggesting a coordinated function for these proteins in the organisation of apical polarity in the parasite.
Collapse
Affiliation(s)
- Benoit Poulin
- Centre for Genetics and Genomics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG2 7UH, UK
| | - Eva-Maria Patzewitz
- Centre for Genetics and Genomics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG2 7UH, UK
| | - Declan Brady
- Centre for Genetics and Genomics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG2 7UH, UK
| | - Olivier Silvie
- INSERM and Université Pierre et Marie Curie, UMR_S 945 “Immunity and infection”, Centre Hospitalier Universitaire Pitié-Salpêtrière, 75013 Paris, France
| | - Megan H. Wright
- Institute of Chemical Biology, Department of Chemistry, Imperial College London, Exhibition Road, London SW7 2AZ, UK
| | - David J. P. Ferguson
- Nuffield Department of Clinical Laboratory Science, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Richard J. Wall
- Centre for Genetics and Genomics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG2 7UH, UK
| | - Sarah Whipple
- Centre for Genetics and Genomics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG2 7UH, UK
| | - David S. Guttery
- Centre for Genetics and Genomics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG2 7UH, UK
- Department of Cancer Studies and Molecular Medicine, University of Leicester, Robert Kilpatrick Building, PO Box 65, Leicester Royal Infirmary, Leicester LE2 7LX, UK
| | - Edward W. Tate
- Institute of Chemical Biology, Department of Chemistry, Imperial College London, Exhibition Road, London SW7 2AZ, UK
| | - Bill Wickstead
- Centre for Genetics and Genomics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG2 7UH, UK
| | - Anthony A. Holder
- Division of Parasitology, MRC National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| | - Rita Tewari
- Centre for Genetics and Genomics, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG2 7UH, UK
| |
Collapse
|
142
|
Abstract
Owing to the absence of antiparasitic vaccines and the constant threat of drug resistance, the development of novel antiparasitic chemotherapies remains of major importance for disease control. A better understanding of drug transport (uptake and efflux), drug metabolism and the identification of drug targets, and mechanisms of drug resistance would facilitate the development of more effective therapies. Here, we focus on malaria and African trypanosomiasis. We review existing drugs and drug development, emphasizing high-throughput genomic and genetic approaches, which hold great promise for elucidating antiparasitic mechanisms. We describe the approaches and technologies that have been influential for each parasite and develop new ideas for future research directions, including mode-of-action studies for drug target deconvolution.
Collapse
Affiliation(s)
- David Horn
- Biological Chemistry & Drug Discovery, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Manoj T. Duraisingh
- Harvard School of Public Health, 665 Huntington Avenue, Building 1, Room 715, Boston, Massachusetts 02115, USA
| |
Collapse
|
143
|
Prommana P, Uthaipibull C, Wongsombat C, Kamchonwongpaisan S, Yuthavong Y, Knuepfer E, Holder AA, Shaw PJ. Inducible knockdown of Plasmodium gene expression using the glmS ribozyme. PLoS One 2013; 8:e73783. [PMID: 24023691 PMCID: PMC3758297 DOI: 10.1371/journal.pone.0073783] [Citation(s) in RCA: 149] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 07/23/2013] [Indexed: 01/07/2023] Open
Abstract
Conventional reverse genetic approaches for study of Plasmodium malaria parasite gene function are limited, or not applicable. Hence, new inducible systems are needed. Here we describe a method to control P. falciparum gene expression in which target genes bearing a glmS ribozyme in the 3' untranslated region are efficiently knocked down in transgenic P. falciparum parasites in response to glucosamine inducer. Using reporter genes, we show that the glmS ribozyme cleaves reporter mRNA in vivo leading to reduction in mRNA expression following glucosamine treatment. Glucosamine-induced ribozyme activation led to efficient reduction of reporter protein, which could be rapidly reversed by removing the inducer. The glmS ribozyme was validated as a reverse-genetic tool by integration into the essential gene and antifolate drug target dihydrofolate reductase-thymidylate synthase (PfDHFR-TS). Glucosamine treatment of transgenic parasites led to rapid and efficient knockdown of PfDHFR-TS mRNA and protein. PfDHFR-TS knockdown led to a growth/arrest mutant phenotype and hypersensitivity to pyrimethamine. The glmS ribozyme may thus be a tool for study of essential genes in P. falciparum and other parasite species amenable to transfection.
Collapse
Affiliation(s)
- Parichat Prommana
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Khlong Nueng, Khlong Luang, Pathum Thani, Thailand
| | - Chairat Uthaipibull
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Khlong Nueng, Khlong Luang, Pathum Thani, Thailand
| | - Chayaphat Wongsombat
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Khlong Nueng, Khlong Luang, Pathum Thani, Thailand
| | - Sumalee Kamchonwongpaisan
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Khlong Nueng, Khlong Luang, Pathum Thani, Thailand
| | - Yongyuth Yuthavong
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Khlong Nueng, Khlong Luang, Pathum Thani, Thailand
| | - Ellen Knuepfer
- Division of Parasitology, MRC National Institute for Medical Research, London, United Kingdom
| | - Anthony A. Holder
- Division of Parasitology, MRC National Institute for Medical Research, London, United Kingdom
| | - Philip J. Shaw
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Khlong Nueng, Khlong Luang, Pathum Thani, Thailand
| |
Collapse
|
144
|
Novel thioredoxin-like proteins are components of a protein complex coating the cortical microtubules of Toxoplasma gondii. EUKARYOTIC CELL 2013; 12:1588-99. [PMID: 23873863 DOI: 10.1128/ec.00082-13] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Microtubules are versatile biopolymers that support numerous vital cellular functions in eukaryotes. The specific properties of microtubules are dependent on distinct microtubule-associated proteins, as the tubulin subunits and microtubule structure are exceptionally conserved. Highly specialized microtubule-containing assemblies are often found in protists, which are rich sources for novel microtubule-associated proteins. A protozoan parasite, Toxoplasma gondii, possesses several distinct tubulin-containing structures, including 22 microtubules closely associated with the cortical membrane. Early ultrastructural studies have shown that the cortical microtubules are heavily decorated with associating proteins. However, little is known about the identities of these proteins. Here, we report the discovery of a novel protein, TrxL1 (for Thioredoxin-Like protein 1), and an associating complex that coats the cortical microtubules. TrxL1 contains a thioredoxin-like fold. To visualize its localization in live parasites by fluorescence, we replaced the endogenous TrxL1 gene with an mEmeraldFP-TrxL1 fusion gene. Structured illumination-based superresolution imaging of this parasite line produced a detailed view of the microtubule cytoskeleton. Despite its stable association with the cortical microtubules in the parasite, TrxL1 does not seem to bind to microtubules directly. Coimmunoprecipitation experiments showed that TrxL1 associates with a protein complex containing SPM1, a previously reported microtubule-associated protein in T. gondii. We also found that SPM1 recruits TrxL1 to the cortical microtubules. Besides SPM1, several other novel proteins are found in the TrxL1-containing complex, including TrxL2, a close homolog of TrxL1. Thus, our results reveal for the first time a microtubule-associated complex in T. gondii.
Collapse
|
145
|
Pino P. From technology to biology: a malaria genetic toolbox for the functional dissection of essential genes. Mol Microbiol 2013; 88:650-4. [DOI: 10.1111/mmi.12232] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2013] [Indexed: 11/28/2022]
Affiliation(s)
- Paco Pino
- Department of Microbiology and Molecular Medicine; CMU; Faculty of Medicine; University of Geneva; Rue Michel-Servet 1; CH-1211; Geneva 4; Switzerland
| |
Collapse
|