1
|
Ngoh IA, Mane K, Manneh J, Bojang F, Jawara AS, Akenji TN, Anong DN, D’Alessandro U, Amambua-Ngwa A. Transcriptome analysis reveals molecular targets of erythrocyte invasion phenotype diversity in natural Plasmodium falciparum isolates from Cameroon. FRONTIERS IN PARASITOLOGY 2024; 3:1370615. [PMID: 39817175 PMCID: PMC11731687 DOI: 10.3389/fpara.2024.1370615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/24/2024] [Indexed: 01/18/2025]
Abstract
Further understanding of the molecular mediators of alternative RBC invasion phenotypes in endemic malaria parasites will support malaria blood-stage vaccine or drug development. This study investigated the prevalence of sialic acid (SA)-dependent and SA-independent RBC invasion pathways in endemic Plasmodium falciparum parasites from Cameroon and compared the schizont stage transcriptomes in these two groups to uncover the wider repertoire of transcriptional variation associated with the use of alternative RBC invasion pathway phenotypes. A two-color flow cytometry-based invasion-inhibition assay against RBCs treated with neuraminidase, trypsin, and chymotrypsin and deep RNA sequencing of schizont stages harvested in the first ex vivo replication cycle in culture were employed in this investigation. RBC invasion phenotypes were determined for 63 isolates from asymptomatic children with uncomplicated malaria. Approximately 80% of the isolates invaded neuraminidase-treated but not chymotrypsin-treated RBCs, representing SA-independent pathways of RBC invasion. The schizont transcriptome profiles of 16 isolates with invasion phenotypes revealed a total of 5,136 gene transcripts, with 85% of isolates predicted at schizont stages. Two distinct transcriptome profile clusters belonging to SA-dependent and SA-independent parasites were obtained by data reduction with principal component analysis. Differential analysis of gene expression between the two clusters implicated, in addition to the well-characterized adhesins, the upregulation of genes encoding proteins mediating merozoite organelle discharges as well as several conserved, virulent, merozoite-associated, and exported proteins. The latter majority have been shown to have structural and physiological relevance to RBC surface remodeling and immune evasion in malaria and thus have potential as anti-invasion targets.
Collapse
Affiliation(s)
- Ines A. Ngoh
- Department of Microbiology and Parasitology, University of Bamenda, Bambili, Cameroon
- Disease Control and Elimination (DCE), Medical Research Council The Gambia Unit at the London School of Hygiene and Tropical Medicine (LSHTM), Fajara, Gambia
| | - Karim Mane
- Disease Control and Elimination (DCE), Medical Research Council The Gambia Unit at the London School of Hygiene and Tropical Medicine (LSHTM), Fajara, Gambia
- Wellcome-Medical Research Council (MRC) Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Jarra Manneh
- Disease Control and Elimination (DCE), Medical Research Council The Gambia Unit at the London School of Hygiene and Tropical Medicine (LSHTM), Fajara, Gambia
| | - Fatoumata Bojang
- Disease Control and Elimination (DCE), Medical Research Council The Gambia Unit at the London School of Hygiene and Tropical Medicine (LSHTM), Fajara, Gambia
| | - Aminata S. Jawara
- Disease Control and Elimination (DCE), Medical Research Council The Gambia Unit at the London School of Hygiene and Tropical Medicine (LSHTM), Fajara, Gambia
| | - Theresia N. Akenji
- Department of Microbiology and Parasitology, University of Bamenda, Bambili, Cameroon
| | - Damian N. Anong
- Department of Microbiology and Parasitology, University of Bamenda, Bambili, Cameroon
| | - Umberto D’Alessandro
- Disease Control and Elimination (DCE), Medical Research Council The Gambia Unit at the London School of Hygiene and Tropical Medicine (LSHTM), Fajara, Gambia
| | - Alfred Amambua-Ngwa
- Disease Control and Elimination (DCE), Medical Research Council The Gambia Unit at the London School of Hygiene and Tropical Medicine (LSHTM), Fajara, Gambia
| |
Collapse
|
2
|
Yarlett N, Jarroll EL, Morada M, Lloyd D. Protists: Eukaryotic single-celled organisms and the functioning of their organelles. Adv Microb Physiol 2024; 84:243-307. [PMID: 38821633 DOI: 10.1016/bs.ampbs.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2024]
Abstract
Organelles are membrane bound structures that compartmentalize biochemical and molecular functions. With improved molecular, biochemical and microscopy tools the diversity and function of protistan organelles has increased in recent years, providing a complex panoply of structure/function relationships. This is particularly noticeable with the description of hydrogenosomes, and the diverse array of structures that followed, having hybrid hydrogenosome/mitochondria attributes. These diverse organelles have lost the major, at one time, definitive components of the mitochondrion (tricarboxylic cycle enzymes and cytochromes), however they all contain the machinery for the assembly of Fe-S clusters, which is the single unifying feature they share. The plasticity of organelles, like the mitochondrion, is therefore evident from its ability to lose its identity as an aerobic energy generating powerhouse while retaining key ancestral functions common to both aerobes and anaerobes. It is interesting to note that the apicoplast, a non-photosynthetic plastid that is present in all apicomplexan protozoa, apart from Cryptosporidium and possibly the gregarines, is also the site of Fe-S cluster assembly proteins. It turns out that in Cryptosporidium proteins involved in Fe-S cluster biosynthesis are localized in the mitochondrial remnant organelle termed the mitosome. Hence, different organisms have solved the same problem of packaging a life-requiring set of reactions in different ways, using different ancestral organelles, discarding what is not needed and keeping what is essential. Don't judge an organelle by its cover, more by the things it does, and always be prepared for surprises.
Collapse
Affiliation(s)
- Nigel Yarlett
- Haskins Laboratories, Pace University, New York, NY, United States; The Department of Chemistry and Physical Sciences, Pace University, New York, NY, United States.
| | - Edward L Jarroll
- Department of Biological Sciences, CUNY-Lehman College, Bronx, NY, United States
| | - Mary Morada
- Haskins Laboratories, Pace University, New York, NY, United States
| | - David Lloyd
- Schools of Biosciences and Engineering, Cardiff University, Wales, United Kingdom
| |
Collapse
|
3
|
Azlan UW, Lau YL, Fong MY. Genetic Diversity and Clustering of the Rhoptry Associated Protein-1 of Plasmodium knowlesi from Peninsular Malaysia and Malaysian Borneo. THE KOREAN JOURNAL OF PARASITOLOGY 2022; 60:393-400. [PMID: 36588415 PMCID: PMC9806503 DOI: 10.3347/kjp.2022.60.6.393] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 11/22/2022] [Indexed: 12/29/2022]
Abstract
Human infection with simian malaria Plasmodium knowlesi is a cause for concern in Southeast Asian countries, especially in Malaysia. A previous study on Peninsular Malaysia P. knowlesi rhoptry associated protein-1 (PkRAP1) gene has discovered the existence of dimorphism. In this study, genetic analysis of PkRAP1 in a larger number of P. knowlesi samples from Malaysian Borneo was conducted. The PkRAP1 of these P. knowlesi isolates was PCR-amplified and sequenced. The newly obtained PkRAP1 gene sequences (n = 34) were combined with those from the previous study (n = 26) and analysed for polymorphism and natural selection. Sequence analysis revealed a higher genetic diversity of PkRAP1 compared to the previous study. Exon II of the gene had higher diversity (π = 0.0172) than exon I (π = 0.0128). The diversity of the total coding region (π = 0.0167) was much higher than those of RAP1 orthologues such as PfRAP-1 (π = 0.0041) and PvRAP1 (π = 0.00088). Z-test results indicated that the gene was under purifying selection. Phylogenetic tree and haplotype network showed distinct clustering of Peninsular Malaysia and Malaysian Borneo PkRAP1 haplotypes. This geographical-based clustering of PkRAP1 haplotypes provides further evidence of the dimorphism of the gene and possible existence of 2 distinct P. knowlesi lineages in Malaysia.
Collapse
|
4
|
Mukherjee S, Nguyen S, Sharma E, Goldberg DE. Maturation and substrate processing topography of the Plasmodium falciparum invasion/egress protease plasmepsin X. Nat Commun 2022; 13:4537. [PMID: 35927261 PMCID: PMC9352755 DOI: 10.1038/s41467-022-32271-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 07/22/2022] [Indexed: 11/08/2022] Open
Abstract
The malaria parasite Plasmodium invades a host erythrocyte, multiplies within a parasitophorous vacuole (PV) and then ruptures the PV and erythrocyte membranes in a process known as egress. Both egress and invasion are controlled by effector proteins discharged from specialized secretory organelles. The aspartic protease plasmepsin X (PM X) regulates activity for many of these effectors, but it is unclear how PM X accesses its diverse substrates that reside in different organelles. PM X also autoprocesses to generate different isoforms. The function of this processing is not understood. We have mapped the self-cleavage sites and have constructed parasites with cleavage site mutations. Surprisingly, a quadruple mutant that remains full-length retains in vitro activity, is trafficked normally, and supports normal egress, invasion and parasite growth. The N-terminal half of the prodomain stays bound to the catalytic domain even after processing and is required for proper intracellular trafficking of PM X. We find that this enzyme cleaves microneme and exoneme substrates before discharge, while the rhoptry substrates that are dependent on PM X activity are cleaved after exoneme discharge into the PV. The data give insight into the temporal, spatial and biochemical control of this unusual but important aspartic protease.
Collapse
Affiliation(s)
- Sumit Mukherjee
- Division of Infectious Diseases, Department of Medicine, and Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Suong Nguyen
- Division of Infectious Diseases, Department of Medicine, and Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Eashan Sharma
- Division of Infectious Diseases, Department of Medicine, and Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel E Goldberg
- Division of Infectious Diseases, Department of Medicine, and Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
5
|
Knudsen AS, Walker MR, Agullet JP, Björnsson KH, Bassi MR, Barfod L. Enhancing neutralization of Plasmodium falciparum using a novel monoclonal antibody against the rhoptry-associated membrane antigen. Sci Rep 2022; 12:3040. [PMID: 35197516 PMCID: PMC8866459 DOI: 10.1038/s41598-022-06921-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 01/31/2022] [Indexed: 11/09/2022] Open
Abstract
The pathogenesis of malaria is associated with blood-stage infection and there is strong evidence that antibodies specific to parasite blood-stage antigens can control parasitemia. This provides a strong rational for applying blood-stage antigen components in a multivalent vaccine, as the induced antibodies in combination can enhance protection. The Plasmodium falciparum rhoptry-associated membrane antigen (PfRAMA) is a promising vaccine target, due to its fundamental role in merozoite invasion and low level of polymorphism. Polyclonal antibodies against PfRAMA are able to inhibit P. falciparum growth and interact synergistically when combined with antibodies against P. falciparum reticulocyte-binding protein 5 (PfRh5) or cysteine-rich protective antigen (PfCyRPA). In this study, we identified a novel PfRAMA-specific mAb with neutralizing activity, which in combination with PfRh5- or PfCyRPA-specific mAbs potentiated the neutralizing effect. By applying phage display technology, we mapped the protective epitope to be in the C-terminal region of PfRAMA. Our results confirmed previous finding of synergy between PfRAMA-, PfRh5- and PfCyRPA-specific antibodies, thereby paving the way of testing these antigens (or fragments of these antigens) in combination to improve the efficacy of blood-stage malaria vaccines. The results emphasize the importance of directing antibody responses towards protective epitopes, as the majority of anti-PfRAMA mAbs were unable to inhibit merozoite invasion of erythrocytes.
Collapse
Affiliation(s)
- Anne S Knudsen
- Department of Immunology and Microbiology, Centre for Medical Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Melanie R Walker
- Department of Immunology and Microbiology, Centre for Medical Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Judit P Agullet
- Department of Immunology and Microbiology, Centre for Medical Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kasper H Björnsson
- Department of Immunology and Microbiology, Centre for Medical Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maria R Bassi
- Department of Immunology and Microbiology, Centre for Medical Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lea Barfod
- Department of Immunology and Microbiology, Centre for Medical Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
6
|
Shakya B, Patel SD, Tani Y, Egan ES. Erythrocyte CD55 mediates the internalization of Plasmodium falciparum parasites. eLife 2021; 10:61516. [PMID: 34028351 PMCID: PMC8184214 DOI: 10.7554/elife.61516] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 05/20/2021] [Indexed: 01/04/2023] Open
Abstract
Invasion of human erythrocytes by the malaria parasite Plasmodium falciparum is a multi-step process. Previously, a forward genetic screen for P. falciparum host factors identified erythrocyte CD55 as essential for invasion, but its specific role and how it interfaces with the other factors that mediate this complex process are unknown. Using CRISPR-Cas9 editing, antibody-based inhibition, and live cell imaging, here we show that CD55 is specifically required for parasite internalization. Pre-invasion kinetics, erythrocyte deformability, and echinocytosis were not influenced by CD55, but entry was inhibited when CD55 was blocked or absent. Visualization of parasites attached to CD55-null erythrocytes points to a role for CD55 in stability and/or progression of the moving junction. Our findings demonstrate that CD55 acts after discharge of the parasite’s rhoptry organelles, and plays a unique role relative to all other invasion receptors. As the requirement for CD55 is strain-transcendent, these results suggest that CD55 or its interacting partners may hold potential as therapeutic targets for malaria.
Collapse
Affiliation(s)
- Bikash Shakya
- Departments of Pediatrics and Microbiology & Immunology, Stanford University School of Medicine, Stanford, United States
| | - Saurabh D Patel
- Zuckerman Institute, Columbia University, New York City, United States
| | | | - Elizabeth S Egan
- Departments of Pediatrics and Microbiology & Immunology, Stanford University School of Medicine, Stanford, United States
| |
Collapse
|
7
|
Cobb DW, Kudyba HM, Villegas A, Hoopmann MR, Baptista RP, Bruton B, Krakowiak M, Moritz RL, Muralidharan V. A redox-active crosslinker reveals an essential and inhibitable oxidative folding network in the endoplasmic reticulum of malaria parasites. PLoS Pathog 2021; 17:e1009293. [PMID: 33534803 PMCID: PMC7886143 DOI: 10.1371/journal.ppat.1009293] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 02/16/2021] [Accepted: 01/07/2021] [Indexed: 12/18/2022] Open
Abstract
Malaria remains a major global health problem, creating a constant need for research to identify druggable weaknesses in P. falciparum biology. As important components of cellular redox biology, members of the Thioredoxin (Trx) superfamily of proteins have received interest as potential drug targets in Apicomplexans. However, the function and essentiality of endoplasmic reticulum (ER)-localized Trx-domain proteins within P. falciparum has not been investigated. We generated conditional mutants of the protein PfJ2—an ER chaperone and member of the Trx superfamily—and show that it is essential for asexual parasite survival. Using a crosslinker specific for redox-active cysteines, we identified PfJ2 substrates as PfPDI8 and PfPDI11, both members of the Trx superfamily as well, which suggests a redox-regulatory role for PfJ2. Knockdown of these PDIs in PfJ2 conditional mutants show that PfPDI11 may not be essential. However, PfPDI8 is required for asexual growth and our data suggest it may work in a complex with PfJ2 and other ER chaperones. Finally, we show that the redox interactions between these Trx-domain proteins in the parasite ER and their substrates are sensitive to small molecule inhibition. Together these data build a model for how Trx-domain proteins in the P. falciparum ER work together to assist protein folding and demonstrate the suitability of ER-localized Trx-domain proteins for antimalarial drug development. One of the leading and persistent causes of childhood mortality in the world is malaria, which is caused by parasites from the genus Plasmodium. Unfortunately, the parasite has developed resistance to all available drugs, making the discovery of new drug targets and potential small molecule inhibitors of essential parasite biology a top priority. A critical pathway required for many different biological processes in the parasite is oxidative folding which requires members of the Thioredoxin (Trx) superfamily of proteins. But we know almost nothing about the function and essentiality of Trx-domain proteins that localize to the endoplasmic reticulum, the origin of the secretory pathway, within P. falciparum. Here we show that a network of Trx-domain containing proteins function together and are essential for parasite survival within human red blood cells. Further, we identify a small molecule inhibitor of the redox activities of these Trx-domain containing proteins. This study demonstrates the suitability of this pathway for future antimalarial drug development.
Collapse
Affiliation(s)
- David W. Cobb
- Department of Cellular Biology, University of Georgia, Georgia, United States of America
- Center for Tropical and Emerging Global Diseases, University of Georgia, Georgia, United States of America
| | - Heather M. Kudyba
- Department of Cellular Biology, University of Georgia, Georgia, United States of America
- Center for Tropical and Emerging Global Diseases, University of Georgia, Georgia, United States of America
| | - Alejandra Villegas
- Department of Cellular Biology, University of Georgia, Georgia, United States of America
- Center for Tropical and Emerging Global Diseases, University of Georgia, Georgia, United States of America
| | - Michael R. Hoopmann
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Rodrigo P. Baptista
- Center for Tropical and Emerging Global Diseases, University of Georgia, Georgia, United States of America
- Institute of Bioinformatics, University of Georgia, Georgia, United States of America
| | - Baylee Bruton
- Department of Cellular Biology, University of Georgia, Georgia, United States of America
| | - Michelle Krakowiak
- Department of Cellular Biology, University of Georgia, Georgia, United States of America
- Center for Tropical and Emerging Global Diseases, University of Georgia, Georgia, United States of America
| | - Robert L. Moritz
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Vasant Muralidharan
- Department of Cellular Biology, University of Georgia, Georgia, United States of America
- Center for Tropical and Emerging Global Diseases, University of Georgia, Georgia, United States of America
- * E-mail:
| |
Collapse
|
8
|
Ito D, Chen JH, Takashima E, Hasegawa T, Otsuki H, Takeo S, Thongkukiatkul A, Han ET, Tsuboi T. Identification of a Novel RAMA/RON3 Rhoptry Protein Complex in Plasmodium falciparum Merozoites. Front Cell Infect Microbiol 2021; 10:605367. [PMID: 33537242 PMCID: PMC7848174 DOI: 10.3389/fcimb.2020.605367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/27/2020] [Indexed: 01/08/2023] Open
Abstract
Malaria causes a half a million deaths annually. The parasite intraerythrocytic lifecycle in the human bloodstream is the major cause of morbidity and mortality. Apical organelles of merozoite stage parasites are involved in the invasion of erythrocytes. A limited number of apical organellar proteins have been identified and characterized for their roles during erythrocyte invasion or subsequent intraerythrocytic parasite development. To expand the repertoire of identified apical organellar proteins we generated a panel of monoclonal antibodies against Plasmodium falciparum schizont-rich parasites and screened the antibodies using immunofluorescence assays. Out of 164 hybridoma lines, 12 clones produced monoclonal antibodies yielding punctate immunofluorescence staining patterns in individual merozoites in late schizonts, suggesting recognition of merozoite apical organelles. Five of the monoclonal antibodies were used to immuno-affinity purify their target antigens and these antigens were identified by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Two known apical organelle protein complexes were identified, the high-molecular mass rhoptry protein complex (PfRhopH1/Clags, PfRhopH2, and PfRhopH3) and the low-molecular mass rhoptry protein complex (rhoptry-associated proteins complex, PfRAP1, and PfRAP2). A novel complex was additionally identified by immunoprecipitation, composed of rhoptry-associated membrane antigen (PfRAMA) and rhoptry neck protein 3 (PfRON3) of P. falciparum. We further identified a region spanning amino acids Q221-E481 within the PfRAMA that may associate with PfRON3 in immature schizonts. Further investigation will be required as to whether PfRAMA and PfRON3 interact directly or indirectly.
Collapse
Affiliation(s)
- Daisuke Ito
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan.,Division of Medical Zoology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Jun-Hu Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Tomoyuki Hasegawa
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Hitoshi Otsuki
- Division of Medical Zoology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Satoru Takeo
- Division of Tropical Diseases and Parasitology, Department of Infectious Diseases, Faculty of Medicine, Kyorin University, Mitaka, Japan
| | | | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| |
Collapse
|
9
|
Detection of the Rhoptry Neck Protein Complex in Plasmodium Sporozoites and Its Contribution to Sporozoite Invasion of Salivary Glands. mSphere 2020; 5:5/4/e00325-20. [PMID: 32817376 PMCID: PMC7440843 DOI: 10.1128/msphere.00325-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Sporozoites are the motile infectious stage that mediates malaria parasite transmission from mosquitoes to the mammalian host. This study addresses the question whether the rhoptry neck protein complex forms and functions in sporozoites, in addition to its role in merozoites. By applying coimmunoprecipitation and sporozoite stage-specific gene knockdown assays, it was demonstrated that RON2, RON4, and RON5 form a complex and are involved in sporozoite invasion of salivary glands via their attachment ability. These findings shed light on the conserved invasion mechanisms among apicomplexan infective stages. In addition, the sporozoite stage-specific gene knockdown system has revealed for the first time in Plasmodium that the RON2 and RON4 interaction reciprocally affects their stability and trafficking to rhoptries. Our study raises the possibility that the RON complex functions during sporozoite maturation as well as migration toward and invasion of target cells. In the Plasmodium life cycle, two infectious stages of parasites, merozoites and sporozoites, share rhoptry and microneme apical structures. A crucial step during merozoite invasion of erythrocytes is the discharge to the host cell membrane of some rhoptry neck proteins as a complex, followed by the formation of a moving junction involving the parasite-secreted protein AMA1 on the parasite membrane. Components of the merozoite rhoptry neck protein complex are also expressed in sporozoites, namely, RON2, RON4, and RON5, suggesting that invasion mechanism elements might be conserved between these infective stages. Recently, we demonstrated that RON2 is required for sporozoite invasion of mosquito salivary gland cells and mammalian hepatocytes, using a sporozoite stage-specific gene knockdown strategy in the rodent malaria parasite model, Plasmodium berghei. Here, we use a coimmunoprecipitation assay and oocyst-derived sporozoite extracts to demonstrate that RON2, RON4, and RON5 also form a complex in sporozoites. The sporozoite stage-specific gene knockdown strategy revealed that both RON4 and RON5 have crucial roles during sporozoite invasion of salivary glands, including a significantly reduced attachment ability required for the onset of gliding. Further analyses indicated that RON2 and RON4 reciprocally affect trafficking to rhoptries in developing sporozoites, while RON5 is independently transported. These findings indicate that the interaction between RON2 and RON4 contributes to their stability and trafficking to rhoptries, in addition to involvement in sporozoite attachment. IMPORTANCE Sporozoites are the motile infectious stage that mediates malaria parasite transmission from mosquitoes to the mammalian host. This study addresses the question whether the rhoptry neck protein complex forms and functions in sporozoites, in addition to its role in merozoites. By applying coimmunoprecipitation and sporozoite stage-specific gene knockdown assays, it was demonstrated that RON2, RON4, and RON5 form a complex and are involved in sporozoite invasion of salivary glands via their attachment ability. These findings shed light on the conserved invasion mechanisms among apicomplexan infective stages. In addition, the sporozoite stage-specific gene knockdown system has revealed for the first time in Plasmodium that the RON2 and RON4 interaction reciprocally affects their stability and trafficking to rhoptries. Our study raises the possibility that the RON complex functions during sporozoite maturation as well as migration toward and invasion of target cells.
Collapse
|
10
|
Florentin A, Cobb DW, Kudyba HM, Muralidharan V. Directing traffic: Chaperone-mediated protein transport in malaria parasites. Cell Microbiol 2020; 22:e13215. [PMID: 32388921 PMCID: PMC7282954 DOI: 10.1111/cmi.13215] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/12/2020] [Accepted: 04/14/2020] [Indexed: 12/16/2022]
Abstract
The ability of eukaryotic parasites from the phylum Apicomplexa to cause devastating diseases is predicated upon their ability to maintain faithful and precise protein trafficking mechanisms. Their parasitic life cycle depends on the trafficking of effector proteins to the infected host cell, transport of proteins to several critical organelles required for survival, as well as transport of parasite and host proteins to the digestive organelles to generate the building blocks for parasite growth. Several recent studies have shed light on the molecular mechanisms parasites utilise to transform the infected host cells, transport proteins to essential metabolic organelles and for biogenesis of organelles required for continuation of their life cycle. Here, we review key pathways of protein transport originating and branching from the endoplasmic reticulum, focusing on the essential roles of chaperones in these processes. Further, we highlight key gaps in our knowledge that prevents us from building a holistic view of protein trafficking in these deadly human pathogens.
Collapse
Affiliation(s)
- Anat Florentin
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA.,Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| | - David W Cobb
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA.,Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| | - Heather M Kudyba
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA.,Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| | - Vasant Muralidharan
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA.,Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
11
|
Sherling ES, Perrin AJ, Knuepfer E, Russell MRG, Collinson LM, Miller LH, Blackman MJ. The Plasmodium falciparum rhoptry bulb protein RAMA plays an essential role in rhoptry neck morphogenesis and host red blood cell invasion. PLoS Pathog 2019; 15:e1008049. [PMID: 31491036 PMCID: PMC6750612 DOI: 10.1371/journal.ppat.1008049] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 09/18/2019] [Accepted: 08/27/2019] [Indexed: 01/08/2023] Open
Abstract
The malaria parasite Plasmodium falciparum invades, replicates within and destroys red blood cells in an asexual blood stage life cycle that is responsible for clinical disease and crucial for parasite propagation. Invasive malaria merozoites possess a characteristic apical complex of secretory organelles that are discharged in a tightly controlled and highly regulated order during merozoite egress and host cell invasion. The most prominent of these organelles, the rhoptries, are twinned, club-shaped structures with a body or bulb region that tapers to a narrow neck as it meets the apical prominence of the merozoite. Different protein populations localise to the rhoptry bulb and neck, but the function of many of these proteins and how they are spatially segregated within the rhoptries is unknown. Using conditional disruption of the gene encoding the only known glycolipid-anchored malarial rhoptry bulb protein, rhoptry-associated membrane antigen (RAMA), we demonstrate that RAMA is indispensable for blood stage parasite survival. Contrary to previous suggestions, RAMA is not required for trafficking of all rhoptry bulb proteins. Instead, RAMA-null parasites display selective mislocalisation of a subset of rhoptry bulb and neck proteins (RONs) and produce dysmorphic rhoptries that lack a distinct neck region. The mutant parasites undergo normal intracellular development and egress but display a fatal defect in invasion and do not induce echinocytosis in target red blood cells. Our results indicate that distinct pathways regulate biogenesis of the two main rhoptry sub-compartments in the malaria parasite. Despite improved control measures over recent decades, malaria is still a considerable health burden across much of the globe. The disease is caused by a single-celled parasite that invades and replicates within host cells. During invasion, the parasite discharges a set of flask-shaped secretory organelles called rhoptries, the contents of which are crucial for invasion as well as for modifications to the host cell that are important for parasite survival. Rhoptry discharge occurs through fusion of the relatively elongated rhoptry neck to the apical surface of the parasite. Different proteins reside within the bulbous rhoptry body and the neck regions, but how these proteins are selectively sent to their correct sub-compartments within the rhoptries and how the rhoptries are formed, is poorly understood. Here we show that a malaria parasite rhoptry bulb protein called rhoptry-associated membrane antigen (RAMA) plays an essential role in rhoptry neck formation and correct trafficking of certain rhoptry neck and bulb proteins. Parasites deficient in RAMA produce malformed rhoptries and–probably as a result—cannot invade host red blood cells. Our work sheds new light on how rhoptries are formed and reveals insights into the mechanism by which the correct sorting of proteins to distinct regions of the rhoptry is regulated.
Collapse
Affiliation(s)
- Emma S. Sherling
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Abigail J. Perrin
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Ellen Knuepfer
- Malaria Parasitology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Matthew R. G. Russell
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Lucy M. Collinson
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Louis H. Miller
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Michael J. Blackman
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
- * E-mail:
| |
Collapse
|
12
|
Oda-Yokouchi Y, Tachibana M, Iriko H, Torii M, Ishino T, Tsuboi T. Plasmodium RON12 localizes to the rhoptry body in sporozoites. Parasitol Int 2018; 68:17-23. [PMID: 30290224 DOI: 10.1016/j.parint.2018.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/18/2018] [Accepted: 10/01/2018] [Indexed: 11/25/2022]
Abstract
Invasion of host cells by apicomplexan parasites is mediated by proteins released from microneme, rhoptry, and dense granule secretory organelles located at the apical end of parasite invasive forms. Microneme secreted proteins establish interactions with host cell receptors and induce exocytosis of the rhoptry organelle. Rhoptry proteins are involved in target cell invasion as well as the formation of the parasitophorous vacuole in which parasites reside during development within the host cell. In Plasmodium merozoites, the rhoptry neck protein (RON) complex consists of RON2, RON4, and RON5, and interacts with apical membrane antigen 1 (AMA1) as a critical structure of the invasion moving junction. PfRON12 is known to localize to the rhoptry neck of merozoites, but its function remains obscure. The roles of RON proteins are largely unknown in sporozoites, the second invasive form of Plasmodium which possesses a conserved apical end secretory structure. Here, we confirm that RON12 is expressed in the rhoptry neck of merozoites in rodent malaria parasites, whereas in contrast we show that RON12 is localized to the rhoptry body in sporozoites. Phenotypic analysis of Plasmodium berghei ron12-disrupted mutants revealed that RON12 is dispensable for sporogony, invasion of mosquito salivary glands and mouse hepatocytes, and development in hepatocytes.
Collapse
Affiliation(s)
- Yuki Oda-Yokouchi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan; Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon Ehime 791-0295, Japan
| | - Mayumi Tachibana
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon Ehime 791-0295, Japan
| | - Hideyuki Iriko
- Division of Global Infectious Diseases, Department of Public Health, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo 654-0142, Japan
| | - Motomi Torii
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon Ehime 791-0295, Japan
| | - Tomoko Ishino
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon Ehime 791-0295, Japan.
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan.
| |
Collapse
|
13
|
Hammoudi PM, Maco B, Dogga SK, Frénal K, Soldati-Favre D. Toxoplasma gondiiTFP1 is an essential transporter family protein critical for microneme maturation and exocytosis. Mol Microbiol 2018; 109:225-244. [DOI: 10.1111/mmi.13981] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 05/04/2018] [Accepted: 05/04/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Pierre-Mehdi Hammoudi
- Department of Microbiology and Molecular Medicine, CMU, Faculty of Medicine; University of Geneva, 1 Rue Michel-Servet; Geneva 1206 Switzerland
| | - Bohumil Maco
- Department of Microbiology and Molecular Medicine, CMU, Faculty of Medicine; University of Geneva, 1 Rue Michel-Servet; Geneva 1206 Switzerland
| | - Sunil Kumar Dogga
- Department of Microbiology and Molecular Medicine, CMU, Faculty of Medicine; University of Geneva, 1 Rue Michel-Servet; Geneva 1206 Switzerland
| | - Karine Frénal
- Department of Microbiology and Molecular Medicine, CMU, Faculty of Medicine; University of Geneva, 1 Rue Michel-Servet; Geneva 1206 Switzerland
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, CMU, Faculty of Medicine; University of Geneva, 1 Rue Michel-Servet; Geneva 1206 Switzerland
| |
Collapse
|
14
|
Lehmann C, Tan MSY, de Vries LE, Russo I, Sanchez MI, Goldberg DE, Deu E. Plasmodium falciparum dipeptidyl aminopeptidase 3 activity is important for efficient erythrocyte invasion by the malaria parasite. PLoS Pathog 2018; 14:e1007031. [PMID: 29768491 PMCID: PMC5973627 DOI: 10.1371/journal.ppat.1007031] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 05/29/2018] [Accepted: 04/15/2018] [Indexed: 11/19/2022] Open
Abstract
Parasite egress from infected erythrocytes and invasion of new red blood cells are essential processes for the exponential asexual replication of the malaria parasite. These two tightly coordinated events take place in less than a minute and are in part regulated and mediated by proteases. Dipeptidyl aminopeptidases (DPAPs) are papain-fold cysteine proteases that cleave dipeptides from the N-terminus of protein substrates. DPAP3 was previously suggested to play an essential role in parasite egress. However, little is known about its enzymatic activity, intracellular localization, or biological function. In this study, we recombinantly expressed DPAP3 and demonstrate that it has indeed dipeptidyl aminopeptidase activity, but contrary to previously studied DPAPs, removal of its internal prodomain is not required for activation. By combining super resolution microscopy, time-lapse fluorescence microscopy, and immunoelectron microscopy, we show that Plasmodium falciparum DPAP3 localizes to apical organelles that are closely associated with the neck of the rhoptries, and from which DPAP3 is secreted immediately before parasite egress. Using a conditional knockout approach coupled to complementation studies with wild type or mutant DPAP3, we show that DPAP3 activity is important for parasite proliferation and critical for efficient red blood cell invasion. We also demonstrate that DPAP3 does not play a role in parasite egress, and that the block in egress phenotype previously reported for DPAP3 inhibitors is due to off target or toxicity effects. Finally, using a flow cytometry assay to differentiate intracellular parasites from extracellular parasites attached to the erythrocyte surface, we show that DPAP3 is involved in the initial attachment of parasites to the red blood cell surface. Overall, this study establishes the presence of a DPAP3-dependent invasion pathway in malaria parasites.
Collapse
Affiliation(s)
- Christine Lehmann
- Chemical Biology Approaches to Malaria Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Michele Ser Ying Tan
- Chemical Biology Approaches to Malaria Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Laura E. de Vries
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ilaria Russo
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Mateo I. Sanchez
- Department of Genetics, Stanford School of Medicine, Stanford, California, United States of America
| | - Daniel E. Goldberg
- Departments of Molecular Microbiology and Medicine, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Edgar Deu
- Chemical Biology Approaches to Malaria Laboratory, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
15
|
Hallée S, Counihan NA, Matthews K, Koning‐Ward TF, Richard D. The malaria parasite
Plasmodium falciparum
Sortilin is essential for merozoite formation and apical complex biogenesis. Cell Microbiol 2018; 20:e12844. [DOI: 10.1111/cmi.12844] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 02/22/2018] [Accepted: 03/17/2018] [Indexed: 12/25/2022]
Affiliation(s)
- Stéphanie Hallée
- Centre de recherche en infectiologieCHU de Québec‐Université Laval Quebec City QC Canada
| | | | - Kathryn Matthews
- School of MedicineDeakin University Waurn Ponds 3216 VIC Australia
| | | | - Dave Richard
- Centre de recherche en infectiologieCHU de Québec‐Université Laval Quebec City QC Canada
| |
Collapse
|
16
|
Hallée S, Thériault C, Gagnon D, Kehrer J, Frischknecht F, Mair GR, Richard D. Identification of a Golgi apparatus protein complex important for the asexual erythrocytic cycle of the malaria parasite Plasmodium falciparum. Cell Microbiol 2018; 20:e12843. [PMID: 29579782 DOI: 10.1111/cmi.12843] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 03/09/2018] [Accepted: 03/09/2018] [Indexed: 12/01/2022]
Abstract
Compared with other eukaryotic cell types, malaria parasites appear to possess a more rudimentary Golgi apparatus being composed of dispersed, unstacked cis and trans-cisternae. Despite playing a central role in the secretory pathway of the parasite, few Plasmodium Golgi resident proteins have been characterised. We had previously identified a new Golgi resident protein of unknown function, which we had named Golgi Protein 1, and now show that it forms a complex with a previously uncharacterised transmembrane protein (Golgi Protein 2, GP2). The Golgi Protein complex localises to the cis-Golgi throughout the erythrocytic cycle and potentially also during the mosquito stages. Analysis of parasite strains where GP1 expression is conditionally repressed and/or the GP2 gene is inactivated reveals that though the Golgi protein complex is not essential at any stage of the parasite life cycle, it is important for optimal asexual development in the blood stages.
Collapse
Affiliation(s)
- Stéphanie Hallée
- Centre de recherche en infectiologie, CHU de Québec-Université Laval, Quebec City, Quebec, Canada
| | - Catherine Thériault
- Centre de recherche en infectiologie, CHU de Québec-Université Laval, Quebec City, Quebec, Canada
| | - Dominic Gagnon
- Centre de recherche en infectiologie, CHU de Québec-Université Laval, Quebec City, Quebec, Canada
| | - Jessica Kehrer
- Integrative Parasitology, Department of Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | - Friedrich Frischknecht
- Integrative Parasitology, Department of Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | - Gunnar R Mair
- Integrative Parasitology, Department of Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | - Dave Richard
- Centre de recherche en infectiologie, CHU de Québec-Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|
17
|
Ghosh S, Chisholm SA, Dans M, Lakkavaram A, Kennedy K, Ralph SA, Counihan NA, de Koning-Ward TF. The cysteine protease dipeptidyl aminopeptidase 3 does not contribute to egress of Plasmodium falciparum from host red blood cells. PLoS One 2018; 13:e0193538. [PMID: 29509772 PMCID: PMC5839547 DOI: 10.1371/journal.pone.0193538] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 02/13/2018] [Indexed: 01/27/2023] Open
Abstract
The ability of Plasmodium parasites to egress from their host red blood cell is critical for the amplification of these parasites in the blood. Previous forward chemical genetic approaches have implicated the subtilisin-like protease (SUB1) and the cysteine protease dipeptidyl aminopeptidase 3 (DPAP3) as key players in egress, with the final step of SUB1 maturation thought to be due to the activity of DPAP3. In this study, we have utilized a reverse genetics approach to engineer transgenic Plasmodium falciparum parasites in which dpap3 expression can be conditionally regulated using the glmS ribozyme based RNA-degrading system. We show that DPAP3, which is expressed in schizont stages and merozoites and localizes to organelles distinct from the micronemes, rhoptries and dense granules, is not required for the trafficking of apical proteins or processing of SUB1 substrates, nor for parasite maturation and egress from red blood cells. Thus, our findings argue against a role for DPAP3 in parasite egress and indicate that the phenotypes observed with DPAP3 inhibitors are due to off-target effects.
Collapse
Affiliation(s)
- Sreejoyee Ghosh
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Scott A. Chisholm
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Madeline Dans
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Asha Lakkavaram
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Kit Kennedy
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Stuart A. Ralph
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | | | | |
Collapse
|
18
|
Evidence that the Plasmodium falciparum Protein Sortilin Potentially Acts as an Escorter for the Trafficking of the Rhoptry-Associated Membrane Antigen to the Rhoptries. mSphere 2018; 3:mSphere00551-17. [PMID: 29299530 PMCID: PMC5750388 DOI: 10.1128/msphere.00551-17] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 12/06/2017] [Indexed: 12/11/2022] Open
Abstract
The rhoptry organelle is critical for the invasion of an erythrocyte by the malaria parasite Plasmodium falciparum. Despite their critical roles, the mechanisms behind their biogenesis are still poorly defined. Our earlier work had suggested that the interaction between the glycosylphosphatidylinositol (GPI)-anchored rhoptry-associated membrane antigen (RAMA) and the soluble rhoptry-associated protein 1 was involved in the transport of the latter from the Golgi apparatus to the rhoptry. However, how this protein complex could interact with the intracellular trafficking machinery was unknown at this stage. Here we show that the P. falciparum homologue of the transmembrane protein sortilin-VPS10 interacts with regions of RAMA that are sufficient to target a fluorescent reporter to the rhoptries. These results suggest that P. falciparum sortilin (PfSortilin) could potentially act as the escorter for the transport of rhoptry-destined cargo. IMPORTANCE The malaria parasite is a massive burden in several parts of the world. Worryingly, the parasite has become resistant to several of the drugs commonly used to treat the disease, and at this time, there is no commercial vaccine. It is therefore critical to identify new targets for the development of antimalarials. To survive in the human body, the malaria parasite needs to invade red blood cells. For this, it uses a variety of effectors stored in organelles forming a structure called the apical complex. The mechanisms behind how the parasite generates the apical complex are poorly understood. In this study, we present evidence that a transmembrane protein called sortilin potentially acts as an escorter to transport proteins from the Golgi apparatus to the rhoptries, a component of the apical complex. Our study provides new insight into the biogenesis of a critical structure of the malaria parasite.
Collapse
|
19
|
Ebrahimzadeh Z, Mukherjee A, Richard D. A map of the subcellular distribution of phosphoinositides in the erythrocytic cycle of the malaria parasite Plasmodium falciparum. Int J Parasitol 2017; 48:13-25. [PMID: 29154995 DOI: 10.1016/j.ijpara.2017.08.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 08/22/2017] [Accepted: 08/31/2017] [Indexed: 12/16/2022]
Abstract
Despite representing a small percentage of the cellular lipids of eukaryotic cells, phosphoinositides (PIPs) are critical in various processes such as intracellular trafficking and signal transduction. Central to their various functions is the differential distribution of PIP species to specific membrane compartments through the actions of kinases, phosphatases and lipases. Despite their importance in the malaria parasite lifecycle, the subcellular distribution of most PIP species in this organism is still unknown. We here localise several species of PIPs throughout the erythrocytic cycle of Plasmodium falciparum. We show that PI3P is mostly found at the apicoplast and the membrane of the food vacuole, that PI4P associates with the Golgi apparatus and the plasma membrane and that PI(4,5)P2, in addition to being detected at the plasma membrane, labels some cavity-like spherical structures. Finally, we show that the elusive PI5P localises to the plasma membrane, the nucleus and potentially to the transitional endoplasmic reticulum (ER). Our map of the subcellular distribution of PIP species in P. falciparum will be a useful tool to shed light on the dynamics of these lipids in this deadly parasite.
Collapse
Affiliation(s)
- Zeinab Ebrahimzadeh
- Centre de recherche en infectiologie, CRCHU de Québec-Université Laval, 2705 Boul. Laurier, Québec, QC G1V 4G2, Canada
| | - Angana Mukherjee
- Centre de recherche en infectiologie, CRCHU de Québec-Université Laval, 2705 Boul. Laurier, Québec, QC G1V 4G2, Canada
| | - Dave Richard
- Centre de recherche en infectiologie, CRCHU de Québec-Université Laval, 2705 Boul. Laurier, Québec, QC G1V 4G2, Canada.
| |
Collapse
|
20
|
Mehlotra RK, Blankenship D, Howes RE, Rakotomanga TA, Ramiranirina B, Ramboarina S, Franchard T, Linger MH, Zikursh-Blood M, Ratsimbasoa AC, Zimmerman PA, Grimberg BT. Long-term in vitro culture of Plasmodium vivax isolates from Madagascar maintained in Saimiri boliviensis blood. Malar J 2017; 16:442. [PMID: 29100506 PMCID: PMC5670718 DOI: 10.1186/s12936-017-2090-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 10/27/2017] [Indexed: 02/06/2023] Open
Abstract
Background Plasmodium vivax is the most prevalent human malaria parasite and is likely to increase proportionally as malaria control efforts more rapidly impact the prevalence of Plasmodium falciparum. Despite the prominence of P. vivax as a major human pathogen, vivax malaria qualifies as a neglected and under-studied tropical disease. Significant challenges bringing P. vivax into the laboratory, particularly the capacity for long-term propagation of well-characterized strains, have limited the study of this parasite’s red blood cell (RBC) invasion mechanism, blood-stage development, gene expression, and genetic manipulation. Methods and results Patient isolates of P. vivax have been collected and cryopreserved in the rural community of Ampasimpotsy, located in the Tsiroanomandidy Health District of Madagascar. Periodic, monthly overland transport of these cryopreserved isolates to the country’s National Malaria Control Programme laboratory in Antananarivo preceded onward sample transfer to laboratories at Case Western Reserve University, USA. There, the P. vivax isolates have been cultured through propagation in the RBCs of Saimiri boliviensis. For the four patient isolates studied to-date, the median time interval between sample collection and in vitro culture has been 454 days (range 166–961 days). The median time in culture, continually documented by light microscopy, has been 159 days; isolate AMP2014.01 was continuously propagated for 233 days. Further studies show that the P. vivax parasites propagated in Saimiri RBCs retain their ability to invade human RBCs, and can be cryopreserved, thawed and successfully returned to productive in vitro culture. Conclusions/significance Long-term culture of P. vivax is possible in the RBCs of Saimiri boliviensis. These studies provide an alternative to propagation of P. vivax in live animals that are becoming more restricted. In vitro culture of P. vivax in Saimiri RBCs provides an opening to stabilize patient isolates, which would serve as precious resources to apply new strategies for investigating the molecular and cellular biology of this important malaria parasite. Electronic supplementary material The online version of this article (10.1186/s12936-017-2090-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rajeev K Mehlotra
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, 44106-4983, USA
| | - D'Arbra Blankenship
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, 44106-4983, USA
| | - Rosalind E Howes
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, 44106-4983, USA.,Oxford Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Tovonahary A Rakotomanga
- National Malaria Control Programme, Ministry of Health, Antananarivo, Madagascar.,Faculty of Sciences, University of Antananarivo, Antananarivo, Madagascar
| | - Brune Ramiranirina
- Faculty of Sciences, University of Antananarivo, Antananarivo, Madagascar
| | - Stephanie Ramboarina
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, 44106-4983, USA.,Faculty of Sciences, University of Antananarivo, Antananarivo, Madagascar
| | - Thierry Franchard
- National Malaria Control Programme, Ministry of Health, Antananarivo, Madagascar.,Faculty of Sciences, University of Antananarivo, Antananarivo, Madagascar
| | - Marlin H Linger
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, 44106-4983, USA
| | - Melinda Zikursh-Blood
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, 44106-4983, USA
| | - Arsène C Ratsimbasoa
- National Malaria Control Programme, Ministry of Health, Antananarivo, Madagascar.,Faculty of Sciences, University of Antananarivo, Antananarivo, Madagascar
| | - Peter A Zimmerman
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, 44106-4983, USA.
| | - Brian T Grimberg
- Center for Global Health and Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, 44106-4983, USA.
| |
Collapse
|
21
|
Thériault C, Richard D. Characterization of a putative Plasmodium falciparum SAC1 phosphoinositide-phosphatase homologue potentially required for survival during the asexual erythrocytic stages. Sci Rep 2017; 7:12710. [PMID: 28983103 PMCID: PMC5629215 DOI: 10.1038/s41598-017-12762-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/15/2017] [Indexed: 12/13/2022] Open
Abstract
Despite marked reductions in morbidity and mortality in the last ten years, malaria still takes a tremendous toll on human populations throughout tropical and sub-tropical regions of the world. The absence of an effective vaccine and resistance to most antimalarial drugs available demonstrate the urgent need for new intervention strategies. Phosphoinositides are a class of lipids with critical roles in numerous processes and their specific subcellular distribution, generated through the action of kinases and phosphatases, define organelle identity in a wide range of eukaryotic cells. Recent studies have highlighted important functions of phosphoinositide kinases in several parts of the Plasmodium lifecycle such as hemoglobin endocytosis and cytokinesis during the erythrocytic stage however, nothing is known with regards to the parasite's putative phosphoinositide phosphatases. We present the identification and initial characterization of a putative homologue of the SAC1 phosphoinositide phosphatase family. Our results show that the protein is expressed throughout the asexual blood stages and that it localises to the endoplasmic reticulum and potentially to the Golgi apparatus. Furthermore, conditional knockdown and knockout studies suggest that a minimal amount of the protein are likely required for survival during the erythrocytic cycle.
Collapse
Affiliation(s)
- Catherine Thériault
- Centre de recherche en infectiologie du CHU de Québec-Université Laval, Quebec City, Quebec, Canada
| | - Dave Richard
- Centre de recherche en infectiologie du CHU de Québec-Université Laval, Quebec City, Quebec, Canada.
| |
Collapse
|
22
|
Deu E. Proteases as antimalarial targets: strategies for genetic, chemical, and therapeutic validation. FEBS J 2017; 284:2604-2628. [PMID: 28599096 PMCID: PMC5575534 DOI: 10.1111/febs.14130] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 04/29/2017] [Accepted: 06/06/2017] [Indexed: 01/17/2023]
Abstract
Malaria is a devastating parasitic disease affecting half of the world's population. The rapid emergence of resistance against new antimalarial drugs, including artemisinin-based therapies, has made the development of drugs with novel mechanisms of action extremely urgent. Proteases are enzymes proven to be well suited for target-based drug development due to our knowledge of their enzymatic mechanisms and active site structures. More importantly, Plasmodium proteases have been shown to be involved in a variety of pathways that are essential for parasite survival. However, pharmacological rather than target-based approaches have dominated the field of antimalarial drug development, in part due to the challenge of robustly validating Plasmodium targets at the genetic level. Fortunately, over the last few years there has been significant progress in the development of efficient genetic methods to modify the parasite, including several conditional approaches. This progress is finally allowing us not only to validate essential genes genetically, but also to study their molecular functions. In this review, I present our current understanding of the biological role proteases play in the malaria parasite life cycle. I also discuss how the recent advances in Plasmodium genetics, the improvement of protease-oriented chemical biology approaches, and the development of malaria-focused pharmacological assays, can be combined to achieve a robust biological, chemical and therapeutic validation of Plasmodium proteases as viable drug targets.
Collapse
Affiliation(s)
- Edgar Deu
- Chemical Biology Approaches to Malaria LaboratoryThe Francis Crick InstituteLondonUK
| |
Collapse
|
23
|
Santos JM, Josling G, Ross P, Joshi P, Orchard L, Campbell T, Schieler A, Cristea IM, Llinás M. Red Blood Cell Invasion by the Malaria Parasite Is Coordinated by the PfAP2-I Transcription Factor. Cell Host Microbe 2017; 21:731-741.e10. [PMID: 28618269 PMCID: PMC5855115 DOI: 10.1016/j.chom.2017.05.006] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 02/16/2017] [Accepted: 05/23/2017] [Indexed: 10/19/2022]
Abstract
Obligate intracellular parasites must efficiently invade host cells in order to mature and be transmitted. For the malaria parasite Plasmodium falciparum, invasion of host red blood cells (RBCs) is essential. Here we describe a parasite-specific transcription factor PfAP2-I, belonging to the Apicomplexan AP2 (ApiAP2) family, that is responsible for regulating the expression of genes involved in RBC invasion. Our genome-wide analysis by ChIP-seq shows that PfAP2-I interacts with a specific DNA motif in the promoters of target genes. Although PfAP2-I contains three AP2 DNA-binding domains, only one is required for binding of the target genes during blood stage development. Furthermore, we find that PfAP2-I associates with several chromatin-associated proteins, including the Plasmodium bromodomain protein PfBDP1 and that complex formation is associated with transcriptional regulation. As a key regulator of red blood cell invasion, PfAP2-I represents a potential new antimalarial therapeutic target.
Collapse
Affiliation(s)
- Joana Mendonca Santos
- Department of Biochemistry and Molecular Biology and Huck Center for Malaria Research, Pennsylvania State University, State College, PA 16802, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Gabrielle Josling
- Department of Biochemistry and Molecular Biology and Huck Center for Malaria Research, Pennsylvania State University, State College, PA 16802, USA
| | - Philipp Ross
- Department of Biochemistry and Molecular Biology and Huck Center for Malaria Research, Pennsylvania State University, State College, PA 16802, USA
| | - Preeti Joshi
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Lindsey Orchard
- Department of Biochemistry and Molecular Biology and Huck Center for Malaria Research, Pennsylvania State University, State College, PA 16802, USA
| | - Tracey Campbell
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Ariel Schieler
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Manuel Llinás
- Department of Biochemistry and Molecular Biology and Huck Center for Malaria Research, Pennsylvania State University, State College, PA 16802, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Chemistry and Huck Center for Infectious Disease Dynamics, Pennsylvania State University, State College, PA 16802, USA.
| |
Collapse
|
24
|
Ghosh S, Kennedy K, Sanders P, Matthews K, Ralph SA, Counihan NA, de Koning-Ward TF. ThePlasmodiumrhoptry associated protein complex is important for parasitophorous vacuole membrane structure and intraerythrocytic parasite growth. Cell Microbiol 2017; 19. [DOI: 10.1111/cmi.12733] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/13/2017] [Accepted: 02/09/2017] [Indexed: 01/15/2023]
Affiliation(s)
- Sreejoyee Ghosh
- School of Medicine; Deakin University; Waurn Ponds Victoria Australia
| | - Kit Kennedy
- Department of Biochemistry and Molecular Biology; Bio21 Molecular Science and Biotechnology Institute; Melbourne Victoria Australia
| | - Paul Sanders
- The Burnet Institute; Melbourne Victoria Australia
| | - Kathryn Matthews
- School of Medicine; Deakin University; Waurn Ponds Victoria Australia
| | - Stuart A. Ralph
- Department of Biochemistry and Molecular Biology; Bio21 Molecular Science and Biotechnology Institute; Melbourne Victoria Australia
| | | | | |
Collapse
|
25
|
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: 69] [Impact Index Per Article: 8.6] [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
|
26
|
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: 0.5] [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
|
27
|
Morse D, Webster W, Kalanon M, Langsley G, McFadden GI. Plasmodium falciparum Rab1A Localizes to Rhoptries in Schizonts. PLoS One 2016; 11:e0158174. [PMID: 27348424 PMCID: PMC4922565 DOI: 10.1371/journal.pone.0158174] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 06/11/2016] [Indexed: 12/17/2022] Open
Abstract
Over-expression of a GFP-PfRab1A fusion protein in Plasmodium falciparum schizonts produces a punctate pattern of fluorescence typical of rhoptries, secretory organelles involved in host cell invasion. The GFP-positive bodies were purified by a combination of differential and density gradient centrifugation and their protein content determined by MS/MS sequencing. Consistent with the GFP rhoptry-like pattern of transgenic parasites, four of the 19 proteins identified have been previously described to be rhoptry-associated and another four are ER or ER-associated proteins. Confirmation that GFP-PfRab1A decorates rhoptries was obtained by its co-localization with Rap1 and Ron4 in late phase schizonts. We conclude that PfRab1A potentially regulates vesicular traffic from the endoplasmic reticulum to the rhoptries in Apicomplexa parasites.
Collapse
Affiliation(s)
- David Morse
- School of BioSciences, University of Melbourne, Melbourne, VIC, 3010, Australia
- * E-mail:
| | - Wesley Webster
- School of BioSciences, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Ming Kalanon
- School of BioSciences, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Gordon Langsley
- Laboratoire de Biologie Cellulaire Comparative des Apicomplexes, Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Descartes, 75014, Paris, France
| | | |
Collapse
|
28
|
Absalon S, Robbins JA, Dvorin JD. An essential malaria protein defines the architecture of blood-stage and transmission-stage parasites. Nat Commun 2016; 7:11449. [PMID: 27121004 PMCID: PMC4853479 DOI: 10.1038/ncomms11449] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 03/29/2016] [Indexed: 11/30/2022] Open
Abstract
Blood-stage replication of the human malaria parasite Plasmodium falciparum occurs via schizogony, wherein daughter parasites are formed by a specialized cytokinesis known as segmentation. Here we identify a parasite protein, which we name P. falciparum Merozoite Organizing Protein (PfMOP), as essential for cytokinesis of blood-stage parasites. We show that, following PfMOP knockdown, parasites undergo incomplete segmentation resulting in a residual agglomerate of partially divided cells. While organelles develop normally, the structural scaffold of daughter parasites, the inner membrane complex (IMC), fails to form in this agglomerate causing flawed segmentation. In PfMOP-deficient gametocytes, the IMC formation defect causes maturation arrest with aberrant morphology and death. Our results provide insight into the mechanisms of replication and maturation of malaria parasites. Blood-stage malaria parasites replicate through a specialised type of cell division known as schizogony. Here, Absalon et al. identify a parasite protein that is essential during schizogony for cytokinesis and formation of the inner membrane complex, the structural scaffold of daughter parasites.
Collapse
Affiliation(s)
- Sabrina Absalon
- Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Jonathan A Robbins
- Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Division of Infectious Diseases, Massachusetts General Hospital/Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | - Jeffrey D Dvorin
- Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
29
|
El Chamy Maluf S, Dal Mas C, Oliveira EB, Melo PM, Carmona AK, Gazarini ML, Hayashi MAF. Inhibition of malaria parasite Plasmodium falciparum development by crotamine, a cell penetrating peptide from the snake venom. Peptides 2016; 78:11-6. [PMID: 26806200 DOI: 10.1016/j.peptides.2016.01.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 01/15/2016] [Accepted: 01/19/2016] [Indexed: 01/24/2023]
Abstract
We show here that crotamine, a polypeptide from the South American rattlesnake venom with cell penetrating and selective anti-fungal and anti-tumoral properties, presents a potent anti-plasmodial activity in culture. Crotamine inhibits the development of the Plasmodium falciparum parasites in a dose-dependent manner [IC50 value of 1.87 μM], and confocal microscopy analysis showed a selective internalization of fluorescent-labeled crotamine into P. falciparum infected erythrocytes, with no detectable fluorescence in uninfected healthy erythrocytes. In addition, similarly to the crotamine cytotoxic effects, the mechanism underlying the anti-plasmodial activity may involve the disruption of parasite acidic compartments H(+) homeostasis. In fact, crotamine promoted a reduction of parasites organelle fluorescence loaded with the lysosomotropic fluorochrome acridine orange, in the same way as previously observed mammalian tumoral cells. Taken together, we show for the first time crotamine not only compromised the metabolism of the P. falciparum, but this toxin also inhibited the parasite growth. Therefore, we suggest this snake polypeptide as a promising lead molecule for the development of potential new molecules, namely peptidomimetics, with selectivity for infected erythrocytes and ability to inhibit the malaria infection by its natural affinity for acid vesicles.
Collapse
Affiliation(s)
- S El Chamy Maluf
- Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - C Dal Mas
- Departamento de Farmacologia, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - E B Oliveira
- Departamento de Bioquímica e Imunologia, Universidade de São Paulo (USP-RP), Ribeirão Preto, Brazil
| | - P M Melo
- Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - A K Carmona
- Departamento de Biofísica, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - M L Gazarini
- Departamento de Biociências, Universidade Federal de São Paulo (UNIFESP), Santos, SP, Brazil.
| | - M A F Hayashi
- Departamento de Farmacologia, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.
| |
Collapse
|
30
|
Tyagi K, Hossain ME, Thakur V, Aggarwal P, Malhotra P, Mohmmed A, Sharma YD. Plasmodium vivax Tryptophan Rich Antigen PvTRAg36.6 Interacts with PvETRAMP and PvTRAg56.6 Interacts with PvMSP7 during Erythrocytic Stages of the Parasite. PLoS One 2016; 11:e0151065. [PMID: 26954579 PMCID: PMC4783080 DOI: 10.1371/journal.pone.0151065] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 02/09/2016] [Indexed: 01/09/2023] Open
Abstract
Plasmodium vivax is most wide spread and a neglected malaria parasite. There is a lack of information on parasite biology of this species. Genome of this parasite encodes for the largest number of tryptophan-rich proteins belonging to ‘Pv-fam-a’ family and some of them are potential drug/vaccine targets but their functional role(s) largely remains unexplored. Using bacterial and yeast two hybrid systems, we have identified the interacting partners for two of the P. vivax tryptophan-rich antigens called PvTRAg36.6 and PvTRAg56.2. The PvTRAg36.6 interacts with early transcribed membrane protein (ETRAMP) of P.vivax. It is apically localized in merozoites but in early stages it is seen in parasite periphery suggesting its likely involvement in parasitophorous vacuole membrane (PVM) development or maintenance. On the other hand, PvTRAg56.2 interacts with P.vivax merozoite surface protein7 (PvMSP7) and is localized on merozoite surface. Co-localization of PvTRAg56.2 with PvMSP1 and its molecular interaction with PvMSP7 probably suggest that, PvTRAg56.2 is part of MSP-complex, and might assist or stabilize the protein complex at the merozoite surface. In conclusion, the PvTRAg proteins have different sub cellular localizations and specific associated functions during intra-erythrocytic developmental cycle.
Collapse
Affiliation(s)
- Kriti Tyagi
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Mohammad Enayet Hossain
- Malaria group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Vandana Thakur
- Malaria group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Praveen Aggarwal
- Department of Emergency Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Pawan Malhotra
- Malaria group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Asif Mohmmed
- Malaria group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- * E-mail: (YDS); (AM)
| | - Yagya Dutta Sharma
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
- * E-mail: (YDS); (AM)
| |
Collapse
|
31
|
Koch M, Baum J. The mechanics of malaria parasite invasion of the human erythrocyte - towards a reassessment of the host cell contribution. Cell Microbiol 2016; 18:319-29. [PMID: 26663815 PMCID: PMC4819681 DOI: 10.1111/cmi.12557] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 11/30/2015] [Accepted: 12/07/2015] [Indexed: 01/15/2023]
Abstract
Despite decades of research, we still know little about the mechanics of Plasmodium host cell invasion. Fundamentally, while the essential or non‐essential nature of different parasite proteins is becoming clearer, their actual function and how each comes together to govern invasion are poorly understood. Furthermore, in recent years an emerging world view is shifting focus away from the parasite actin–myosin motor being the sole force responsible for entry to an appreciation of host cell dynamics and forces and their contribution to the process. In this review, we discuss merozoite invasion of the erythrocyte, focusing on the complex set of pre‐invasion events and how these might prime the red cell to facilitate invasion. While traditionally parasite interactions at this stage have been viewed simplistically as mediating adhesion only, recent work makes it apparent that by interacting with a number of host receptors and signalling pathways, combined with secretion of parasite‐derived lipid material, that the merozoite may initiate cytoskeletal re‐arrangements and biophysical changes in the erythrocyte that greatly reduce energy barriers for entry. Seen in this light Plasmodium invasion may well turn out to be a balance between host and parasite forces, much like that of other pathogen infection mechanisms.
Collapse
Affiliation(s)
- Marion Koch
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, SW7 2AZ, UK
| | - Jake Baum
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, SW7 2AZ, UK
| |
Collapse
|
32
|
Kaderi Kibria KM, Rawat K, Klinger CM, Datta G, Panchal M, Singh S, Iyer GR, Kaur I, Sharma V, Dacks JB, Mohmmed A, Malhotra P. A role for adaptor protein complex 1 in protein targeting to rhoptry organelles in Plasmodium falciparum. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:699-710. [PMID: 25573429 DOI: 10.1016/j.bbamcr.2014.12.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 12/12/2014] [Accepted: 12/25/2014] [Indexed: 12/20/2022]
Abstract
The human malaria parasite Plasmodium falciparum possesses sophisticated systems of protein secretion to modulate host cell invasion and remodeling. In the present study, we provide insights into the function of the AP-1 complex in P. falciparum. We utilized GFP fusion constructs for live cell imaging, as well as fixed parasites in immunofluorescence analysis, to study adaptor protein mu1 (Pfμ1) mediated protein trafficking in P. falciparum. In trophozoites Pfμ1 showed similar dynamic localization to that of several Golgi/ER markers, indicating Golgi/ER localization. Treatment of transgenic parasites with Brefeldin A altered the localization of Golgi-associated Pfμ1, supporting the localization studies. Co-localization studies showed considerable overlap of Pfμ1 with the resident rhoptry proteins, rhoptry associated protein 1 (RAP1) and Cytoadherence linked asexual gene 3.1 (Clag3.1) in schizont stage. Immunoprecipitation experiments with Pfμ1 and PfRAP1 revealed an interaction, which may be mediated through an intermediate transmembrane cargo receptor. A specific role for Pfμ1 in trafficking was suggested by treatment with AlF4, which resulted in a shift to a predominantly ER-associated compartment and consequent decrease in co-localization with the Golgi marker GRASP. Together, these results suggest a role for the AP-1 complex in rhoptry protein trafficking in P. falciparum.
Collapse
Affiliation(s)
- K M Kaderi Kibria
- Malaria Research Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Khushboo Rawat
- Malaria Research Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Christen M Klinger
- Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada
| | - Gaurav Datta
- Malaria Research Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Manoj Panchal
- Malaria Research Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Shailja Singh
- Malaria Research Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Gayatri R Iyer
- Malaria Research Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Inderjeet Kaur
- Malaria Research Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Veena Sharma
- Department of Bioscience and Biotechnology, Banasthali University, Banasthali-304022, Rajasthan, India
| | - Joel B Dacks
- Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada.
| | - Asif Mohmmed
- Malaria Research Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India.
| | - Pawan Malhotra
- Malaria Research Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India.
| |
Collapse
|
33
|
Kumar P, Tripathi A, Ranjan R, Halbert J, Gilberger T, Doerig C, Sharma P. Regulation of Plasmodium falciparum development by calcium-dependent protein kinase 7 (PfCDPK7). J Biol Chem 2014; 289:20386-95. [PMID: 24895132 DOI: 10.1074/jbc.m114.561670] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Second messengers such as phosphoinositides and calcium are known to control diverse processes involved in the development of malaria parasites. However, the underlying molecular mechanisms and pathways need to be unraveled, which may be achieved by understanding the regulation of effectors of these second messengers. Calcium-dependent protein kinase (CDPK) family members regulate diverse parasitic processes. Because CDPKs are absent from the host, these kinases are considered as potential drug targets. We have dissected the function of an atypical CDPK from Plasmodium falciparum, PfCDPK7. The domain architecture of PfCDPK7 is very different from that of other CDPKs; it has a pleckstrin homology domain adjacent to the kinase domain and two calcium-binding EF-hands at its N terminus. We demonstrate that PfCDPK7 interacts with PI(4,5)P2 via its pleckstrin homology domain, which may guide its subcellular localization. Disruption of PfCDPK7 caused a marked reduction in the growth of the blood stage parasites, as maturation of rings to trophozoites was markedly stalled. In addition, parasite proliferation was significantly attenuated. These findings shed light on an important role for PfCDPK7 in the erythrocytic asexual cycle of malaria parasites.
Collapse
Affiliation(s)
- Praveen Kumar
- From the Eukaryotic Gene Expression Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Anuj Tripathi
- From the Eukaryotic Gene Expression Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Ravikant Ranjan
- From the Eukaryotic Gene Expression Laboratory, National Institute of Immunology, New Delhi 110067, India
| | - Jean Halbert
- the Inserm-EPFL Joint Laboratory, Global Health Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Tim Gilberger
- the Department of Pathology and Molecular Medicine, M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4L8, Canada, the Bernhard-Nocht-Institute for Tropical Medicine, 20359 Hamburg, Germany, and
| | - Christian Doerig
- the Inserm-EPFL Joint Laboratory, Global Health Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland, the Department of Microbiology, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| | - Pushkar Sharma
- From the Eukaryotic Gene Expression Laboratory, National Institute of Immunology, New Delhi 110067, India,
| |
Collapse
|
34
|
RON5 is critical for organization and function of the Toxoplasma moving junction complex. PLoS Pathog 2014; 10:e1004025. [PMID: 24651769 PMCID: PMC3961375 DOI: 10.1371/journal.ppat.1004025] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 02/10/2014] [Indexed: 11/19/2022] Open
Abstract
Apicomplexans facilitate host cell invasion through formation of a tight-junction interface between parasite and host plasma membranes called the moving junction (MJ). A complex of the rhoptry neck proteins RONs 2/4/5/8 localize to the MJ during invasion where they are believed to provide a stable anchoring point for host penetration. During the initiation of invasion, the preformed MJ RON complex is injected into the host cell where RON2 spans the host plasma membrane while RONs 4/5/8 localize to its cytosolic face. While much attention has been directed toward an AMA1-RON2 interaction supposed to occur outside the cell, little is known about the functions of the MJ RONs positioned inside the host cell. Here we provide a detailed analysis of RON5 to resolve outstanding questions about MJ complex organization, assembly and function during invasion. Using a conditional knockdown approach, we show loss of RON5 results in complete degradation of RON2 and mistargeting of RON4 within the parasite secretory pathway, demonstrating that RON5 plays a key role in organization of the MJ RON complex. While RON8 is unaffected by knockdown of RON5, these parasites are unable to invade new host cells, providing the first genetic demonstration that RON5 plays a critical role in host cell penetration. Although invasion is not required for injection of rhoptry effectors into the host cytosol, parasites lacking RON5 also fail to form evacuoles suggesting an intact MJ complex is a prerequisite for secretion of rhoptry bulb contents. Additionally, while the MJ has been suggested to function in egress, disruption of the MJ complex by RON5 depletion does not impact this process. Finally, functional complementation of our conditional RON5 mutant reveals that while proteolytic separation of RON5 N- and C-terminal fragments is dispensable, a portion of the C-terminal domain is critical for RON2 stability and function in invasion. Toxoplasma and related apicomplexan parasites are obligate intracellular pathogens that actively invade their host cells, creating a specialized vacuole within which the parasite is able to replicate. Invasion involves the establishment of a tight-junction interface between host and parasite membranes called the moving junction (MJ) through which the parasite actively penetrates the host. At the onset of invasion, a protein complex composed of RONs 2/4/5/8 is injected from specialized parasite secretory organelles called rhoptries into the host membrane. Following secretion, this RON complex localizes to the MJ throughout the invasion event and is thought to be the basis for this tight-junction. In this study, we utilize a conditional knockdown of RON5 to show that this MJ component, present at the cytosolic face of the host membrane during penetration, is crucial for invasion and for MJ complex organization. In particular, loss of RON5 results in degradation of RON2 and mistargeting of RON4 in the parasite, effectively ablating the MJ complex. We exploit this knockdown strain to evaluate RON5 processing and identify regions of the protein that are necessary for organizing the complex. Our findings demonstrate the key role of RON5 in facilitating apicomplexan host invasion and disease.
Collapse
|
35
|
Curtidor H, Patiño LC, Arévalo-Pinzón G, Vanegas M, Patarroyo ME, Patarroyo MA. Plasmodium falciparum rhoptry neck protein 5 peptides bind to human red blood cells and inhibit parasite invasion. Peptides 2014; 53:210-7. [PMID: 23932940 DOI: 10.1016/j.peptides.2013.07.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 07/31/2013] [Accepted: 07/31/2013] [Indexed: 10/26/2022]
Abstract
Plasmodium falciparum malaria parasite invasion of erythrocytes is an essential step in host infection and the proteins involved in such invasion are the main target in developing an antimalarial vaccine. Secretory organelle-derived proteins (micronemal AMA1 protein and the RON2, 4, and 5 rhoptry neck proteins) have been recently described as components of moving junction complex formation allowing merozoites to move into a newly created parasitophorous vacuole. This study led to identifying RON5 regions involved in binding to human erythrocytes by using a highly robust, sensitive and specific receptor-ligand interaction assay; it is further shown that the RON5 protein remains highly conserved throughout different parasite strains. It is shown that the binding peptide-erythrocyte interaction is saturable and sensitive to chymotrypsin and trypsin. Invasion inhibition assays using erythrocyte binding peptides showed that the RON5-erythrocyte interaction could be critical for merozoite invasion of erythrocytes. This work provides evidence (for the first time) suggesting a fundamental role for RON5 in erythrocyte invasion.
Collapse
Affiliation(s)
- Hernando Curtidor
- Universidad de la Sabana, Km. 7, Autopista Norte, Bogotá, Colombia; Universidad del Rosario, Calle 14 No. 6-25, Bogotá, Colombia; Fundación Instituto de Inmunología de Colombia, Carrera 50 No. 26-20, Bogotá, Colombia.
| | - Liliana C Patiño
- Universidad del Rosario, Calle 14 No. 6-25, Bogotá, Colombia; Fundación Instituto de Inmunología de Colombia, Carrera 50 No. 26-20, Bogotá, Colombia
| | - Gabriela Arévalo-Pinzón
- Universidad del Rosario, Calle 14 No. 6-25, Bogotá, Colombia; Fundación Instituto de Inmunología de Colombia, Carrera 50 No. 26-20, Bogotá, Colombia
| | - Magnolia Vanegas
- Universidad del Rosario, Calle 14 No. 6-25, Bogotá, Colombia; Fundación Instituto de Inmunología de Colombia, Carrera 50 No. 26-20, Bogotá, Colombia
| | - Manuel E Patarroyo
- Fundación Instituto de Inmunología de Colombia, Carrera 50 No. 26-20, Bogotá, Colombia; Universidad Nacional de Colombia, Carrera 45 No. 26-85, Bogotá, Colombia
| | - Manuel A Patarroyo
- Universidad del Rosario, Calle 14 No. 6-25, Bogotá, Colombia; Fundación Instituto de Inmunología de Colombia, Carrera 50 No. 26-20, Bogotá, Colombia
| |
Collapse
|
36
|
Molecular cloning and characterization of NcROP2Fam-1, a member of the ROP2 family of rhoptry proteins in Neospora caninum that is targeted by antibodies neutralizing host cell invasion in vitro. Parasitology 2014; 140:1033-50. [PMID: 23743240 DOI: 10.1017/s0031182013000383] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Recent publications demonstrated that a fragment of a Neospora caninum ROP2 family member antigen represents a promising vaccine candidate. We here report on the cloning of the cDNA encoding this protein, N. caninum ROP2 family member 1 (NcROP2Fam-1), its molecular characterization and localization. The protein possesses the hallmarks of ROP2 family members and is apparently devoid of catalytic activity. NcROP2Fam-1 is synthesized as a pre-pro-protein that is matured to 2 proteins of 49 and 55 kDa that localize to rhoptry bulbs. Upon invasion the protein is associated with the nascent parasitophorous vacuole membrane (PVM), evacuoles surrounding the host cell nucleus and, in some instances, the surface of intracellular parasites. Staining was also observed within the cyst wall of 'cysts' produced in vitro. Interestingly, NcROP2Fam-1 was also detected on the surface of extracellular parasites entering the host cells and antibodies directed against NcROP2Fam-1-specific peptides partially neutralized invasion in vitro. We conclude that, in spite of the general belief that ROP2 family proteins are intracellular antigens, NcROP2Fam-1 can also be considered as an extracellular antigen, a property that should be taken into account in further experiments employing ROP2 family proteins as vaccines.
Collapse
|
37
|
Tomavo S, Slomianny C, Meissner M, Carruthers VB. Protein trafficking through the endosomal system prepares intracellular parasites for a home invasion. PLoS Pathog 2013; 9:e1003629. [PMID: 24204248 PMCID: PMC3812028 DOI: 10.1371/journal.ppat.1003629] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Toxoplasma (toxoplasmosis) and Plasmodium (malaria) use unique secretory organelles for migration, cell invasion, manipulation of host cell functions, and cell egress. In particular, the apical secretory micronemes and rhoptries of apicomplexan parasites are essential for successful host infection. New findings reveal that the contents of these organelles, which are transported through the endoplasmic reticulum (ER) and Golgi, also require the parasite endosome-like system to access their respective organelles. In this review, we discuss recent findings that demonstrate that these parasites reduced their endosomal system and modified classical regulators of this pathway for the biogenesis of apical organelles.
Collapse
Affiliation(s)
- Stanislas Tomavo
- Center for Infection and Immunity of Lille, CNRS UMR 8204, INSERM U 1019, Institut Pasteur de Lille, Université Lille Nord de France, Lille, France
- * E-mail:
| | - Christian Slomianny
- Laboratory of Cell Physiology, INSERM U 1003, Université Lille Nord de France, Villeneuve d'Ascq, Lille, France
| | - Markus Meissner
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Vern B. Carruthers
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| |
Collapse
|
38
|
Comparative genomic analysis of multi-subunit tethering complexes demonstrates an ancient pan-eukaryotic complement and sculpting in Apicomplexa. PLoS One 2013; 8:e76278. [PMID: 24086721 PMCID: PMC3785458 DOI: 10.1371/journal.pone.0076278] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 08/22/2013] [Indexed: 11/19/2022] Open
Abstract
Apicomplexa are obligate intracellular parasites that cause tremendous disease burden world-wide. They utilize a set of specialized secretory organelles in their invasive process that require delivery of components for their biogenesis and function, yet the precise mechanisms underpinning such processes remain unclear. One set of potentially important components is the multi-subunit tethering complexes (MTCs), factors increasingly implicated in all aspects of vesicle-target interactions. Prompted by the results of previous studies indicating a loss of membrane trafficking factors in Apicomplexa, we undertook a bioinformatic analysis of MTC conservation. Building on knowledge of the ancient presence of most MTC proteins, we demonstrate the near complete retention of MTCs in the newly available genomes for Guillardiatheta and Bigelowiellanatans. The latter is a key taxonomic sampling point as a basal sister taxa to the group including Apicomplexa. We also demonstrate an ancient origin of the CORVET complex subunits Vps8 and Vps3, as well as the TRAPPII subunit Tca17. Having established that the lineage leading to Apicomplexa did at one point possess the complete eukaryotic complement of MTC components, we undertook a deeper taxonomic investigation in twelve apicomplexan genomes. We observed excellent conservation of the VpsC core of the HOPS and CORVET complexes, as well as the core TRAPP subunits, but sparse conservation of TRAPPII, COG, Dsl1, and HOPS/CORVET-specific subunits. However, those subunits that we did identify appear to be expressed with similar patterns to the fully conserved MTC proteins, suggesting that they may function as minimal complexes or with analogous partners. Strikingly, we failed to identify any subunits of the exocyst complex in all twelve apicomplexan genomes, as well as the dinoflagellate Perkinsus marinus. Overall, we demonstrate reduction of MTCs in Apicomplexa and their ancestors, consistent with modification during, and possibly pre-dating, the move from free-living marine algae to deadly human parasites.
Collapse
|
39
|
Klinger CM, Nisbet RE, Ouologuem DT, Roos DS, Dacks JB. Cryptic organelle homology in apicomplexan parasites: insights from evolutionary cell biology. Curr Opin Microbiol 2013; 16:424-31. [PMID: 23932202 PMCID: PMC4513074 DOI: 10.1016/j.mib.2013.07.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 07/18/2013] [Accepted: 07/23/2013] [Indexed: 12/21/2022]
Abstract
The economic and clinical significance of apicomplexan parasites drives interest in their many evolutionary novelties. Distinctive intracellular organelles play key roles in parasite motility, invasion, metabolism, and replication, and understanding their relationship with the organelles of better-studied eukaryotic systems suggests potential targets for therapeutic intervention. Recent work has demonstrated divergent aspects of canonical eukaryotic components in the Apicomplexa, including Golgi bodies and mitochondria. The apicoplast is a relict plastid of secondary endosymbiotic origin, harboring metabolic pathways distinct from those of host species. The inner membrane complex (IMC) is derived from the cortical alveoli defining the superphylum Alveolata, but in apicomplexans functions in parasite motility and replication. Micronemes and rhoptries are associated with establishment of the intracellular niche, and define the apical complex for which the phylum is named. Morphological, cell biological and molecular evidence strongly suggest that these organelles are derived from the endocytic pathway.
Collapse
Affiliation(s)
| | - R. Ellen Nisbet
- Department of Biochemistry, Cambridge University, Cambridge UK and School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA, Australia
- Department of Biology, University of Pennsylvania, Philadelphia PA USA
| | | | | | - Joel B. Dacks
- Department of Cell Biology, University of Alberta, Edmonton AB, Canada
| |
Collapse
|
40
|
Zhang Y, Jiang N, Lu H, Hou N, Piao X, Cai P, Yin J, Wahlgren M, Chen Q. Proteomic analysis of Plasmodium falciparum schizonts reveals heparin-binding merozoite proteins. J Proteome Res 2013; 12:2185-93. [PMID: 23566259 DOI: 10.1021/pr400038j] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The malaria parasite Plasmodium falciparum utilizes host glycosaminoglycans (GAGs) as receptors for erythrocyte invasion and intravascular sequestration. Heparin and heparan sulfate (HS) are GAGs which can block erythrocyte invasion of the P. falciparum merozoite, albeit the molecular mechanisms remain poorly understood. Characterization of these heparin-binding merozoite proteins and key ligands in the host-parasite interplay will lead to a better understanding of the mechanism of erythrocyte invasion by malaria parasites. Here, schizont-derived proteins that bind heparin were enriched by affinity chromatography, and 6062 peptides from 811 P. falciparum-derived proteins were identified by two-dimensional liquid chromatography-mass spectrometry (LC/LC-MS/MS). The proteins were categorized into 14 functional groups ranging from pathogenesis, protein catabolic process to signal transduction. Proteins with predominant peptide counts were found to mainly originate from the rhoptry organelle of merozoites and the parasitized erythrocyte membrane. The profile of the heparin/HS-binding proteome of P. falciparum suggests they have important functions in the biology of the parasite.
Collapse
Affiliation(s)
- Yan Zhang
- Key Laboratory of Zoonosis, Ministry of Education, Jilin University, Xian Da Lu 5333, Changchun 130062, China
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Counihan NA, Kalanon M, Coppel RL, de Koning-Ward TF. Plasmodium rhoptry proteins: why order is important. Trends Parasitol 2013; 29:228-36. [PMID: 23570755 DOI: 10.1016/j.pt.2013.03.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 02/28/2013] [Accepted: 03/04/2013] [Indexed: 11/26/2022]
Abstract
Apicomplexan parasites, including the Plasmodium species that cause malaria, contain three unusual apical secretory organelles (micronemes, rhoptries, and dense granules) that are required for the infection of new host cells. Because of their specialized nature, the majority of proteins secreted from these organelles are unique to Apicomplexans and are consequently poorly characterized. Although rhoptry proteins of Plasmodium have been implicated in events central to invasion, there is growing evidence to suggest that proteins originating from this organelle play key roles downstream of parasite entry into the host cell. Here we discuss recent work that has advanced our knowledge of rhoptry protein trafficking and function, and highlight areas of research that require further investigation.
Collapse
|
42
|
Chakrabarti R, Rawat PS, Cooke BM, Coppel RL, Patankar S. Cellular effects of curcumin on Plasmodium falciparum include disruption of microtubules. PLoS One 2013; 8:e57302. [PMID: 23505424 PMCID: PMC3591428 DOI: 10.1371/journal.pone.0057302] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 01/21/2013] [Indexed: 12/17/2022] Open
Abstract
Curcumin has been widely investigated for its myriad cellular effects resulting in reduced proliferation of various eukaryotic cells including cancer cells and the human malaria parasite Plasmodium falciparum. Studies with human cancer cell lines HT-29, Caco-2, and MCF-7 suggest that curcumin can bind to tubulin and induce alterations in microtubule structure. Based on this finding, we investigated whether curcumin has any effect on P. falciparum microtubules, considering that mammalian and parasite tubulin are 83% identical. IC50 of curcumin was found to be 5 µM as compared to 20 µM reported before. Immunofluorescence images of parasites treated with 5 or 20 µM curcumin showed a concentration-dependent effect on parasite microtubules resulting in diffuse staining contrasting with the discrete hemispindles and subpellicular microtubules observed in untreated parasites. The effect on P. falciparum microtubules was evident only in the second cycle for both concentrations tested. This diffuse pattern of tubulin fluorescence in curcumin treated parasites was similar to the effect of a microtubule destabilizing drug vinblastine on P. falciparum. Molecular docking predicted the binding site of curcumin at the interface of alpha and beta tubulin, similar to another destabilizing drug colchicine. Data from predicted drug binding is supported by results from drug combination assays showing antagonistic interactions between curcumin and colchicine, sharing a similar binding site, and additive/synergistic interactions of curcumin with paclitaxel and vinblastine, having different binding sites. This evidence suggests that cellular effects of curcumin are at least, in part, due to its perturbing effect on P. falciparum microtubules. The action of curcumin, both direct and indirect, on P. falciparum microtubules is discussed.
Collapse
Affiliation(s)
- Rimi Chakrabarti
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IITB), Mumbai, India
- Department of Microbiology, Monash University, Melbourne, Victoria, Australia
- IITB-Monash Research Academy, IIT Bombay, Mumbai, Maharashtra, India
| | - Parkash S. Rawat
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IITB), Mumbai, India
| | - Brian M. Cooke
- Department of Microbiology, Monash University, Melbourne, Victoria, Australia
| | - Ross L. Coppel
- Department of Microbiology, Monash University, Melbourne, Victoria, Australia
| | - Swati Patankar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay (IITB), Mumbai, India
| |
Collapse
|
43
|
Ghoneim AM. Trafficking of Plasmodium falciparum chimeric rhoptry protein with Brefeldin A. Folia Parasitol (Praha) 2013; 60:75-8. [DOI: 10.14411/fp.2013.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
44
|
Cowman AF, Berry D, Baum J. The cellular and molecular basis for malaria parasite invasion of the human red blood cell. ACTA ACUST UNITED AC 2013; 198:961-71. [PMID: 22986493 PMCID: PMC3444787 DOI: 10.1083/jcb.201206112] [Citation(s) in RCA: 232] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Malaria is a major disease of humans caused by protozoan parasites from the genus Plasmodium. It has a complex life cycle; however, asexual parasite infection within the blood stream is responsible for all disease pathology. This stage is initiated when merozoites, the free invasive blood-stage form, invade circulating erythrocytes. Although invasion is rapid, it is the only time of the life cycle when the parasite is directly exposed to the host immune system. Significant effort has, therefore, focused on identifying the proteins involved and understanding the underlying mechanisms behind merozoite invasion into the protected niche inside the human erythrocyte.
Collapse
Affiliation(s)
- Alan F Cowman
- The Walter and Eliza Hall Institute of Medical Research, University of Melbourne, Victoria, 3052, Australia.
| | | | | |
Collapse
|
45
|
Deponte M, Hoppe HC, Lee MC, Maier AG, Richard D, Rug M, Spielmann T, Przyborski JM. Wherever I may roam: Protein and membrane trafficking in P. falciparum-infected red blood cells. Mol Biochem Parasitol 2012; 186:95-116. [DOI: 10.1016/j.molbiopara.2012.09.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 09/21/2012] [Accepted: 09/24/2012] [Indexed: 11/27/2022]
|
46
|
Zuccala ES, Gout AM, Dekiwadia C, Marapana DS, Angrisano F, Turnbull L, Riglar DT, Rogers KL, Whitchurch CB, Ralph SA, Speed TP, Baum J. Subcompartmentalisation of proteins in the rhoptries correlates with ordered events of erythrocyte invasion by the blood stage malaria parasite. PLoS One 2012; 7:e46160. [PMID: 23049965 PMCID: PMC3458004 DOI: 10.1371/journal.pone.0046160] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2012] [Accepted: 08/27/2012] [Indexed: 11/18/2022] Open
Abstract
Host cell infection by apicomplexan parasites plays an essential role in lifecycle progression for these obligate intracellular pathogens. For most species, including the etiological agents of malaria and toxoplasmosis, infection requires active host-cell invasion dependent on formation of a tight junction – the organising interface between parasite and host cell during entry. Formation of this structure is not, however, shared across all Apicomplexa or indeed all parasite lifecycle stages. Here, using an in silico integrative genomic search and endogenous gene-tagging strategy, we sought to characterise proteins that function specifically during junction-dependent invasion, a class of proteins we term invasins to distinguish them from adhesins that function in species specific host-cell recognition. High-definition imaging of tagged Plasmodium falciparum invasins localised proteins to multiple cellular compartments of the blood stage merozoite. This includes several that localise to distinct subcompartments within the rhoptries. While originating from the same organelle, however, each has very different dynamics during invasion. Apical Sushi Protein and Rhoptry Neck protein 2 release early, following the junction, whilst a novel rhoptry protein PFF0645c releases only after invasion is complete. This supports the idea that organisation of proteins within a secretory organelle determines the order and destination of protein secretion and provides a localisation-based classification strategy for predicting invasin function during apicomplexan parasite invasion.
Collapse
Affiliation(s)
- Elizabeth S. Zuccala
- Infection and Immunity, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Alexander M. Gout
- Bioinformatics Divisions, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Chaitali Dekiwadia
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Danushka S. Marapana
- Infection and Immunity, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Fiona Angrisano
- Infection and Immunity, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Lynne Turnbull
- The ithree Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| | - David T. Riglar
- Infection and Immunity, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Kelly L. Rogers
- Imaging Facility, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Cynthia B. Whitchurch
- The ithree Institute, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Stuart A. Ralph
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Terence P. Speed
- Bioinformatics Divisions, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Jake Baum
- Infection and Immunity, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- * E-mail:
| |
Collapse
|
47
|
Goldston AM, Powell RR, Temesvari LA. Sink or swim: lipid rafts in parasite pathogenesis. Trends Parasitol 2012; 28:417-26. [PMID: 22906512 DOI: 10.1016/j.pt.2012.07.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 07/16/2012] [Accepted: 07/17/2012] [Indexed: 12/26/2022]
Abstract
Lipid rafts, sterol- and sphingolipid-rich membrane microdomains, have been extensively studied in mammalian cells. Recently, lipid rafts have been shown to control virulence in a variety of parasites including Entamoeba histolytica, Giardia intestinalis, Leishmania spp., Plasmodium spp., Toxoplasma gondii, and Trypanosoma spp. Parasite rafts regulate adhesion to host and invasion, and parasite adhesion molecules often localize to rafts. Parasite rafts also control vesicle trafficking, motility, and cell signaling. Parasites disrupt host cell rafts; the dysregulation of host membrane function facilitates the establishment of infection and evasion of the host immune system. Discerning the mechanism by which lipid rafts regulate parasite pathogenesis is essential to our understanding of virulence. Such insight may guide the development of new drugs for disease management.
Collapse
Affiliation(s)
- Amanda M Goldston
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA
| | | | | |
Collapse
|
48
|
Spielmann T, Montagna GN, Hecht L, Matuschewski K. Molecular make-up of the Plasmodium parasitophorous vacuolar membrane. Int J Med Microbiol 2012; 302:179-86. [PMID: 22898489 DOI: 10.1016/j.ijmm.2012.07.011] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Plasmodium, the causative agent of malaria, is an obligate, intracellular, eukaryotic cell that invades, replicates, and differentiates within hepatocytes and erythrocytes. Inside a host cell, a second membrane delineates the developing pathogen in addition to the parasite plasma membrane, resulting in a distinct cellular compartment, termed parasitophorous vacuole (PV). The PV membrane (PVM) constitutes the parasite-host cell interface and is likely central to nutrient acquisition, host cell remodeling, waste disposal, environmental sensing, and protection from innate defense. Over the past two decades, a number of parasite-encoded PVM proteins have been identified. They include multigene families and protein complexes, such as early-transcribed membrane proteins (ETRAMPs) and the Plasmodium translocon for exported proteins (PTEX). Nearly all Plasmodium PVM proteins are restricted to this genus and display transient and stage-specific expression. Here, we provide an overview of the PVM proteins of Plasmodium blood and liver stages. Biochemical and experimental genetics data suggest that some PVM proteins are ideal targets for novel anti-malarial intervention strategies.
Collapse
Affiliation(s)
- Tobias Spielmann
- Department of Molecular Parasitology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.
| | | | | | | |
Collapse
|
49
|
Static and dynamic imaging of erythrocyte invasion and early intra-erythrocytic development in Plasmodium falciparum. Methods Mol Biol 2012; 923:269-80. [PMID: 22990784 DOI: 10.1007/978-1-62703-026-7_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cellular imaging has reemerged in recent years as a powerful approach to provide researchers with a direct measure of essential molecular events in a cell's life, ranging in scale from broad morphological observations of whole cells to intricate single molecule imaging. When combined with quantitative image analysis, the available imaging techniques can act as a critical means to confirm hypotheses, drive the formation of new theories or provide accurate determination of protein localization at subcellular and nanometer scales. Here, we describe two methodological approaches for imaging the transient step of malaria parasite invasion of the human erythrocyte. When applied to image the most virulent human malaria parasite, Plasmodium falciparum, the first approach, using live time-lapse wide-field microscopy, allows the capture of transient events during invasion and postinvasion intra-erythrocytic development, while the second, using immunofluorescence assay (IFA) of fixed samples, allows high-definition exploration of parasite architecture on multiple platforms.
Collapse
|
50
|
Hajagos BE, Turetzky JM, Peng ED, Cheng SJ, Ryan CM, Souda P, Whitelegge JP, Lebrun M, Dubremetz JF, Bradley PJ. Molecular dissection of novel trafficking and processing of the Toxoplasma gondii rhoptry metalloprotease toxolysin-1. Traffic 2011; 13:292-304. [PMID: 22035499 DOI: 10.1111/j.1600-0854.2011.01308.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 10/27/2011] [Accepted: 10/28/2011] [Indexed: 11/30/2022]
Abstract
Toxoplasma gondii utilizes specialized secretory organelles called rhoptries to invade and hijack its host cell. Many rhoptry proteins are proteolytically processed at a highly conserved SΦXE site to remove organellar targeting sequences that may also affect protein activity. We have studied the trafficking and biogenesis of a secreted rhoptry metalloprotease with homology to insulysin that we named toxolysin-1 (TLN1). Through genetic ablation and molecular dissection of TLN1, we have identified the smallest rhoptry targeting domain yet reported and expanded the consensus sequence of the rhoptry pro-domain cleavage site. In addition to removal of its pro-domain, TLN1 undergoes a C-terminal cleavage event that occurs at a processing site not previously seen in Toxoplasma rhoptry proteins. While pro-domain cleavage occurs in the nascent rhoptries, processing of the C-terminal region precedes commitment to rhoptry targeting, suggesting that it is mediated by a different maturase, and we have identified residues critical for proteolysis. We have additionally shown that both pieces of TLN1 associate in a detergent-resistant complex, formation of which is necessary for trafficking of the C-terminal portion to the rhoptries. Together, these studies reveal novel processing and trafficking events that are present in the protein constituents of this unusual secretory organelle.
Collapse
Affiliation(s)
- Bettina E Hajagos
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095-1489, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|