1
|
Kals E, Kals M, Lees RA, Introini V, Kemp A, Silvester E, Collins CR, Umrekar T, Kotar J, Cicuta P, Rayner JC. Application of optical tweezer technology reveals that PfEBA and PfRH ligands, not PfMSP1, play a central role in Plasmodium falciparum merozoite-erythrocyte attachment. PLoS Pathog 2024; 20:e1012041. [PMID: 39312588 PMCID: PMC11449297 DOI: 10.1371/journal.ppat.1012041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 10/03/2024] [Accepted: 08/05/2024] [Indexed: 09/25/2024] Open
Abstract
Malaria pathogenesis and parasite multiplication depend on the ability of Plasmodium merozoites to invade human erythrocytes. Invasion is a complex multi-step process involving multiple parasite proteins which can differ between species and has been most extensively studied in P. falciparum. However, dissecting the precise role of individual proteins has to date been limited by the availability of quantifiable phenotypic assays. In this study, we apply a new approach to assigning function to invasion proteins by using optical tweezers to directly manipulate recently egressed P. falciparum merozoites and erythrocytes and quantify the strength of attachment between them, as well as the frequency with which such attachments occur. Using a range of inhibitors, antibodies, and genetically modified strains including some generated specifically for this work, we quantitated the contribution of individual P. falciparum proteins to these merozoite-erythrocyte attachment interactions. Conditional deletion of the major P. falciparum merozoite surface protein PfMSP1, long thought to play a central role in initial attachment, had no impact on the force needed to pull merozoites and erythrocytes apart, whereas interventions that disrupted the function of several members of the EBA-175 like Antigen (PfEBA) family and Reticulocyte Binding Protein Homologue (PfRH) invasion ligand families did have a significant negative impact on attachment. Deletion of individual PfEBA and PfRH ligands reinforced the known redundancy within these families, with the deletion of some ligands impacting detachment force while others did not. By comparing over 4000 individual merozoite-erythrocyte interactions in a range of conditions and strains, we establish that the PfEBA/PfRH families play a central role in P. falciparum merozoite attachment, not the major merozoite surface protein PfMSP1.
Collapse
Affiliation(s)
- Emma Kals
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Morten Kals
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Rebecca A. Lees
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck, University of London, London, United Kingdom
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Viola Introini
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
- EMBL Barcelona, Barcelona, Spain
| | - Alison Kemp
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Eleanor Silvester
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Christine R. Collins
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Trishant Umrekar
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck, University of London, London, United Kingdom
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Jurij Kotar
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Pietro Cicuta
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Julian C. Rayner
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
2
|
Morano AA, Ali I, Dvorin JD. Elucidating the spatio-temporal dynamics of the Plasmodium falciparum basal complex. PLoS Pathog 2024; 20:e1012265. [PMID: 38829893 PMCID: PMC11175456 DOI: 10.1371/journal.ppat.1012265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 06/13/2024] [Accepted: 05/16/2024] [Indexed: 06/05/2024] Open
Abstract
Asexual replication of Plasmodium falciparum occurs via schizogony, wherein 16-36 daughter cells are produced within the parasite during one semi-synchronized cytokinetic event. Schizogony requires a divergent contractile ring structure known as the basal complex. Our lab has previously identified PfMyoJ (PF3D7_1229800) and PfSLACR (PF3D7_0214700) as basal complex proteins recruited midway through segmentation. Using ultrastructure expansion microscopy, we localized both proteins to a novel basal complex subcompartment. While both colocalize with the basal complex protein PfCINCH upon recruitment, they form a separate, more basal subcompartment termed the posterior cup during contraction. We also show that PfSLACR is recruited to the basal complex prior to PfMyoJ, and that both proteins are removed unevenly as segmentation concludes. Using live-cell microscopy, we show that actin dynamics are dispensable for basal complex formation, expansion, and contraction. We then show that EF-hand containing P. falciparum Centrin 2 partially localizes to this posterior cup of the basal complex and that it is essential for growth and replication, with variable defects in basal complex contraction and synchrony. Finally, we demonstrate that free intracellular calcium is necessary but not sufficient for basal complex contraction in P. falciparum. Thus, we demonstrate dynamic spatial compartmentalization of the Plasmodium falciparum basal complex, identify an additional basal complex protein, and begin to elucidate the unique mechanism of contraction utilized by P. falciparum, opening the door for further exploration of Apicomplexan cellular division.
Collapse
Affiliation(s)
- Alexander A. Morano
- Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| | - Ilzat Ali
- Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| | - Jeffrey D. Dvorin
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, United States of America
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
3
|
Nayeri T, Sarvi S, Daryani A. Effective factors in the pathogenesis of Toxoplasmagondii. Heliyon 2024; 10:e31558. [PMID: 38818168 PMCID: PMC11137575 DOI: 10.1016/j.heliyon.2024.e31558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 05/11/2024] [Accepted: 05/17/2024] [Indexed: 06/01/2024] Open
Abstract
Toxoplasma gondii (T. gondii) is a cosmopolitan protozoan parasite in humans and animals. It infects about 30 % of the human population worldwide and causes potentially fatal diseases in immunocompromised hosts and neonates. For this study, five English-language databases (ScienceDirect, ProQuest, Web of Science, PubMed, and Scopus) and the internet search engine Google Scholar were searched. This review was accomplished to draw a global perspective of what is known about the pathogenesis of T. gondii and various factors affecting it. Virulence and immune responses can influence the mechanisms of parasite pathogenesis and these factors are in turn influenced by other factors. In addition to the host's genetic background, the type of Toxoplasma strain, the routes of transmission of infection, the number of passages, and different phases of parasite life affect virulence. The identification of virulence factors of the parasite could provide promising insights into the pathogenesis of this parasite. The results of this study can be an incentive to conduct more intensive research to design and develop new anti-Toxoplasma agents (drugs and vaccines) to treat or prevent this infection. In addition, further studies are needed to better understand the key agents in the pathogenesis of T. gondii.
Collapse
Affiliation(s)
- Tooran Nayeri
- Infectious and Tropical Diseases Research Center, Dezful University of Medical Sciences, Dezful, Iran
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shahabeddin Sarvi
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ahmad Daryani
- Department of Parasitology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
4
|
Gurung P, McGee JP, Dvorin JD. PfCAP-H is essential for assembly of condensin I complex and karyokinesis during asexual proliferation of Plasmodium falciparum. mBio 2024; 15:e0285023. [PMID: 38564676 PMCID: PMC11078010 DOI: 10.1128/mbio.02850-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 03/12/2024] [Indexed: 04/04/2024] Open
Abstract
Condensin I is a pentameric complex that regulates the mitotic chromosome assembly in eukaryotes. The kleisin subunit CAP-H of the condensin I complex acts as a linchpin to maintain the structural integrity and loading of this complex on mitotic chromosomes. This complex is present in all eukaryotes and has recently been identified in Plasmodium spp. However, how this complex is assembled and whether the kleisin subunit is critical for this complex in these parasites are yet to be explored. To examine the role of PfCAP-H during cell division within erythrocytes, we generated an inducible PfCAP-H knockout parasite. We find that PfCAP-H is dynamically expressed during mitosis with the peak expression at the metaphase plate. PfCAP-H interacts with PfCAP-G and is a non-SMC member of the condensin I complex. Notably, the absence of PfCAP-H does not alter the expression of PfCAP-G but affects its localization at the mitotic chromosomes. While mitotic spindle assembly is intact in PfCAP-H-deficient parasites, duplicated centrosomes remain clustered over the mass of unsegmented nuclei with failed karyokinesis. This failure leads to the formation of an abnormal nuclear mass, while cytokinesis occurs normally. Altogether, our data suggest that PfCAP-H plays a crucial role in maintaining the structural integrity of the condensin I complex on the mitotic chromosomes and is essential for the asexual development of malarial parasites. IMPORTANCE Mitosis is a fundamental process for Plasmodium parasites, which plays a vital role in their survival within two distinct hosts-human and Anopheles mosquitoes. Despite its great significance, our comprehension of mitosis and its regulation remains limited. In eukaryotes, mitosis is regulated by one of the pivotal complexes known as condensin complexes. The condensin complexes are responsible for chromosome condensation, ensuring the faithful distribution of genetic material to daughter cells. While condensin complexes have recently been identified in Plasmodium spp., our understanding of how this complex is assembled and its precise functions during the blood stage development of Plasmodium falciparum remains largely unexplored. In this study, we investigate the role of a central protein, PfCAP-H, during the blood stage development of P. falciparum. Our findings reveal that PfCAP-H is essential and plays a pivotal role in upholding the structure of condensin I and facilitating karyokinesis.
Collapse
Affiliation(s)
- Pratima Gurung
- Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - James P. McGee
- Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Jeffrey D. Dvorin
- Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Gurung P, McGee JP, Dvorin JD. PfCAP-H is essential for assembly of condensin I complex and karyokinesis during asexual proliferation of Plasmodium falciparum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582160. [PMID: 38464281 PMCID: PMC10925219 DOI: 10.1101/2024.02.26.582160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Condensin I is a pentameric complex that regulates the mitotic chromosome assembly in eukaryotes. The kleisin subunit CAP-H of the condensin I complex acts as a linchpin to maintain the structural integrity and loading of this complex on mitotic chromosomes. This complex is present in all eukaryotes and has recently been identified in Plasmodium spp. However, how this complex is assembled and whether the kleisin subunit is critical for this complex in these parasites is yet to be explored. To examine the role of PfCAP-H during cell division within erythrocytes, we generated an inducible PfCAP-H knockout parasite. We find that PfCAP-H is dynamically expressed during mitosis with the peak expression at the metaphase plate. PfCAP-H interacts with PfCAP-G and is a non-SMC member of the condensin I complex. Notably, the absence of PfCAP-H does not alter the expression of PfCAP-G but affects its localization at the mitotic chromosomes. While mitotic spindle assembly is intact in PfCAP-H deficient parasites, duplicated centrosomes remain clustered over the mass of unsegmented nuclei with failed karyokinesis. This failure leads to the formation of an abnormal nuclear mass, while cytokinesis occurs normally. Altogether, our data suggest that PfCAP-H plays a crucial role in maintaining the structural integrity of the condensin I complex on the mitotic chromosomes and is essential for the asexual development of malarial parasites.
Collapse
Affiliation(s)
- Pratima Gurung
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, M.A
- Department of Pediatrics, Harvard Medical School, Boston, M.A
| | - James P. McGee
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, M.A
| | - Jeffrey D. Dvorin
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, M.A
- Department of Pediatrics, Harvard Medical School, Boston, M.A
| |
Collapse
|
6
|
Liffner B, Cepeda Diaz AK, Blauwkamp J, Anaguano D, Frolich S, Muralidharan V, Wilson DW, Dvorin JD, Absalon S. Atlas of Plasmodium falciparum intraerythrocytic development using expansion microscopy. eLife 2023; 12:RP88088. [PMID: 38108809 PMCID: PMC10727503 DOI: 10.7554/elife.88088] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023] Open
Abstract
Apicomplexan parasites exhibit tremendous diversity in much of their fundamental cell biology, but study of these organisms using light microscopy is often hindered by their small size. Ultrastructural expansion microscopy (U-ExM) is a microscopy preparation method that physically expands the sample by ~4.5×. Here, we apply U-ExM to the human malaria parasite Plasmodium falciparum during the asexual blood stage of its lifecycle to understand how this parasite is organized in three dimensions. Using a combination of dye-conjugated reagents and immunostaining, we have cataloged 13 different P. falciparum structures or organelles across the intraerythrocytic development of this parasite and made multiple observations about fundamental parasite cell biology. We describe that the outer centriolar plaque and its associated proteins anchor the nucleus to the parasite plasma membrane during mitosis. Furthermore, the rhoptries, Golgi, basal complex, and inner membrane complex, which form around this anchoring site while nuclei are still dividing, are concurrently segregated and maintain an association to the outer centriolar plaque until the start of segmentation. We also show that the mitochondrion and apicoplast undergo sequential fission events while maintaining an association with the outer centriolar plaque during cytokinesis. Collectively, this study represents the most detailed ultrastructural analysis of P. falciparum during its intraerythrocytic development to date and sheds light on multiple poorly understood aspects of its organelle biogenesis and fundamental cell biology.
Collapse
Affiliation(s)
- Benjamin Liffner
- Department of Pharmacology and Toxicology, Indiana University School of MedicineIndianapolisUnited States
| | - Ana Karla Cepeda Diaz
- Biological and Biomedical Sciences, Harvard Medical SchoolBostonUnited States
- Division of Infectious Diseases, Boston Children’s HospitalBostonUnited States
| | - James Blauwkamp
- Department of Pharmacology and Toxicology, Indiana University School of MedicineIndianapolisUnited States
| | - David Anaguano
- Center for Tropical and Emerging Global Diseases, University of GeorgiaAthensUnited States
- Department of Cellular Biology, Franklin College of Arts and Sciences, University of GeorgiaAthensUnited States
| | - Sonja Frolich
- Research Centre for Infectious Diseases, School of Biological Sciences, University of AdelaideAdelaideAustralia
- Institute for Photonics and Advanced Sensing, University of AdelaideAdelaideAustralia
| | - Vasant Muralidharan
- Center for Tropical and Emerging Global Diseases, University of GeorgiaAthensUnited States
- Department of Cellular Biology, Franklin College of Arts and Sciences, University of GeorgiaAthensUnited States
| | - Danny W Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, University of AdelaideAdelaideAustralia
- Institute for Photonics and Advanced Sensing, University of AdelaideAdelaideAustralia
- Burnet Institute, 85 Commercial RoadMelbourneAustralia
| | - Jeffrey D Dvorin
- Division of Infectious Diseases, Boston Children’s HospitalBostonUnited States
- Department of Pediatrics, Harvard Medical SchoolBostonUnited States
| | - Sabrina Absalon
- Department of Pharmacology and Toxicology, Indiana University School of MedicineIndianapolisUnited States
| |
Collapse
|
7
|
Ong HW, de Silva C, Avalani K, Kwarcinski F, Mansfield CR, Chirgwin M, Truong A, Derbyshire ER, Zutshi R, Drewry DH. Characterization of 2,4-Dianilinopyrimidines Against Five P. falciparum Kinases PfARK1, PfARK3, PfNEK3, PfPK9, and PfPKB. ACS Med Chem Lett 2023; 14:1774-1784. [PMID: 38116430 PMCID: PMC10726455 DOI: 10.1021/acsmedchemlett.3c00354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 12/21/2023] Open
Abstract
Plasmodium kinases are increasingly recognized as potential novel antiplasmodial targets for the treatment of malaria, but only a small subset of these kinases have had structure-activity relationship (SAR) campaigns reported. Herein we report the discovery of CZC-54252 (1) as an inhibitor of five P. falciparum kinases PfARK1, PfARK3, PfNEK3, PfPK9, and PfPKB. 39 analogues were evaluated against all five kinases to establish SAR at three regions of the kinase active site. Nanomolar inhibitors of each kinase were discovered. We identified common and divergent SAR trends across all five kinases, highlighting substituents in each region that improve potency and selectivity for each kinase. Potent analogues were evaluated against the P. falciparum blood stage. Eight submicromolar inhibitors were discovered, of which 37 demonstrated potent antiplasmodial activity (EC50 = 0.16 μM). Our results provide an understanding of features needed to inhibit each individual kinase and lay groundwork for future optimization efforts toward novel antimalarials.
Collapse
Affiliation(s)
- Han Wee Ong
- Structural
Genomics Consortium and Division of Chemical Biology and Medicinal
Chemistry, Eshelman School of Pharmacy,
University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Chandi de Silva
- Luceome
Biotechnologies, LLC, 1665 East 18th Street, Suite 106, Tucson, Arizona 85719, United States
| | - Krisha Avalani
- Luceome
Biotechnologies, LLC, 1665 East 18th Street, Suite 106, Tucson, Arizona 85719, United States
| | - Frank Kwarcinski
- Luceome
Biotechnologies, LLC, 1665 East 18th Street, Suite 106, Tucson, Arizona 85719, United States
| | - Christopher R. Mansfield
- Department
of Molecular Genetics and Microbiology, Duke University Medical Center, 213 Research Drive, Durham, North Carolina 27710, United States
| | - Michael Chirgwin
- Department
of Chemistry, Duke University, 124 Science Drive, Durham, North Carolina 27708, United States
| | - Anna Truong
- Department
of Chemistry, Duke University, 124 Science Drive, Durham, North Carolina 27708, United States
| | - Emily R. Derbyshire
- Department
of Molecular Genetics and Microbiology, Duke University Medical Center, 213 Research Drive, Durham, North Carolina 27710, United States
- Department
of Chemistry, Duke University, 124 Science Drive, Durham, North Carolina 27708, United States
| | - Reena Zutshi
- Luceome
Biotechnologies, LLC, 1665 East 18th Street, Suite 106, Tucson, Arizona 85719, United States
| | - David H. Drewry
- Structural
Genomics Consortium and Division of Chemical Biology and Medicinal
Chemistry, Eshelman School of Pharmacy,
University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Lineberger
Comprehensive Cancer Center, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
8
|
Cepeda Diaz AK, Rudlaff RM, Farringer M, Dvorin JD. Essential function of alveolin PfIMC1g in the Plasmodium falciparum asexual blood stage. mBio 2023; 14:e0150723. [PMID: 37712738 PMCID: PMC10653860 DOI: 10.1128/mbio.01507-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 07/24/2023] [Indexed: 09/16/2023] Open
Abstract
IMPORTANCE Infection by the Plasmodium falciparum parasite is responsible for the most severe form of human malaria. The asexual blood stage of the parasite, which occurs inside human red blood cells, is responsible for the symptoms of malaria and is the target of most antimalarial drugs. Plasmodium spp. rely on their highly divergent cytoskeletal structures to scaffold their cell division, sustain the mechanical stress of invasion, and survive in both the human bloodstream and the mosquito. We investigate the function of a class of divergent intermediate filament-like proteins called alveolins in the clinically important blood stage. The functional role of individual alveolins in Plasmodium remains poorly understood due to pleiotropic effects of gene knockouts and redundancy among alveolins. We evaluate the localization and essentiality of the four asexual-stage alveolins and find that PfIMC1g and PfIMC1c are essential. Furthermore, we demonstrate that PfIMC1g is critical for survival of the parasite post-invasion.
Collapse
Affiliation(s)
- Ana Karla Cepeda Diaz
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, USA
- Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, USA
| | - Rachel M. Rudlaff
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, USA
- Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, USA
| | - Madeline Farringer
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, USA
- Biological Sciences in Public Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Jeffrey D. Dvorin
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Liffner B, Cepeda Diaz AK, Blauwkamp J, Anaguano D, Frölich S, Muralidharan V, Wilson DW, Dvorin J, Absalon S. Atlas of Plasmodium falciparum intraerythrocytic development using expansion microscopy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.22.533773. [PMID: 36993606 PMCID: PMC10055389 DOI: 10.1101/2023.03.22.533773] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Apicomplexan parasites exhibit tremendous diversity in much of their fundamental cell biology, but study of these organisms using light microscopy is often hindered by their small size. Ultrastructural expansion microscopy (U-ExM) is a microscopy preparation method that physically expands the sample ~4.5x. Here, we apply U-ExM to the human malaria parasite Plasmodium falciparum during the asexual blood stage of its lifecycle to understand how this parasite is organized in three-dimensions. Using a combination of dye-conjugated reagents and immunostaining, we have catalogued 13 different P. falciparum structures or organelles across the intraerythrocytic development of this parasite and made multiple observations about fundamental parasite cell biology. We describe that the outer centriolar plaque and its associated proteins anchor the nucleus to the parasite plasma membrane during mitosis. Furthermore, the rhoptries, Golgi, basal complex, and inner membrane complex, which form around this anchoring site while nuclei are still dividing, are concurrently segregated and maintain an association to the outer centriolar plaque until the start of segmentation. We also show that the mitochondrion and apicoplast undergo sequential fission events while maintaining an association with the outer centriolar plaque during cytokinesis. Collectively, this study represents the most detailed ultrastructural analysis of P. falciparum during its intraerythrocytic development to date, and sheds light on multiple poorly understood aspects of its organelle biogenesis and fundamental cell biology.
Collapse
Affiliation(s)
- Benjamin Liffner
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ana Karla Cepeda Diaz
- Biological and Biomedical Sciences, Harvard Medical School, Boston MA, USA
- Division of Infectious Diseases, Boston Children’s Hospital, Boston MA, USA
| | - James Blauwkamp
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David Anaguano
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
- Department of Cellular Biology, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Sonja Frölich
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Vasant Muralidharan
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
- Department of Cellular Biology, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Danny W. Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
- Institute for Photonics and Advanced Sensing, University of Adelaide, Adelaide, SA, Australia
- Burnet Institute, 85 Commercial Road, Melbourne, VIC, Australia
| | - Jeffrey Dvorin
- Division of Infectious Diseases, Boston Children’s Hospital, Boston MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Sabrina Absalon
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
10
|
Kundu P, Naskar D, McKie SJ, Dass S, Kanjee U, Introini V, Ferreira MU, Cicuta P, Duraisingh M, Deane JE, Rayner JC. The structure of a Plasmodium vivax Tryptophan Rich Antigen domain suggests a lipid binding function for a pan-Plasmodium multi-gene family. Nat Commun 2023; 14:5703. [PMID: 37709739 PMCID: PMC10502043 DOI: 10.1038/s41467-023-40885-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 08/10/2023] [Indexed: 09/16/2023] Open
Abstract
Tryptophan Rich Antigens (TRAgs) are encoded by a multi-gene family found in all Plasmodium species, but are significantly expanded in P. vivax and closely related parasites. We show that multiple P. vivax TRAgs are expressed on the merozoite surface and that one, PVP01_0000100 binds red blood cells with a strong preference for reticulocytes. Using X-ray crystallography, we solved the structure of the PVP01_0000100 C-terminal tryptophan rich domain, which defines the TRAg family, revealing a three-helical bundle that is conserved across Plasmodium and has structural homology with lipid-binding BAR domains involved in membrane remodelling. Biochemical assays confirm that the PVP01_0000100 C-terminal domain has lipid binding activity with preference for sulfatide, a glycosphingolipid present in the outer leaflet of plasma membranes. Deletion of the putative orthologue in P. knowlesi, PKNH_1300500, impacts invasion in reticulocytes, suggesting a role during this essential process. Together, this work defines an emerging molecular function for the Plasmodium TRAg family.
Collapse
Affiliation(s)
- Prasun Kundu
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Deboki Naskar
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Shannon J McKie
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Sheena Dass
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Usheer Kanjee
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Viola Introini
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
| | - Marcelo U Ferreira
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Global Health and Tropical Medicine, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Institute of Hygiene and Tropical Medicine, NOVA University of Lisbon, Lisbon, Portugal
| | - Pietro Cicuta
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
| | - Manoj Duraisingh
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Janet E Deane
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK.
| | - Julian C Rayner
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
11
|
Morano AA, Rudlaff RM, Dvorin JD. A PPP-type pseudophosphatase is required for the maintenance of basal complex integrity in Plasmodium falciparum. Nat Commun 2023; 14:3916. [PMID: 37400439 DOI: 10.1038/s41467-023-39435-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 06/13/2023] [Indexed: 07/05/2023] Open
Abstract
During its asexual blood stage, P. falciparum replicates via schizogony, wherein dozens of daughter cells are formed within a single parent. The basal complex, a contractile ring that separates daughter cells, is critical for schizogony. In this study, we identify a Plasmodium basal complex protein essential for basal complex maintenance. Using multiple microscopy techniques, we demonstrate that PfPPP8 is required for uniform basal complex expansion and maintenance of its integrity. We characterize PfPPP8 as the founding member of a novel family of pseudophosphatases with homologs in other Apicomplexan parasites. By co-immunoprecipitation, we identify two additional new basal complex proteins. We characterize the unique temporal localizations of these new basal complex proteins (late-arriving) and of PfPPP8 (early-departing). In this work, we identify a novel basal complex protein, determine its specific role in segmentation, identify a new pseudophosphatase family, and establish that the P. falciparum basal complex is a dynamic structure.
Collapse
Affiliation(s)
- Alexander A Morano
- Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, 02115, USA
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Rachel M Rudlaff
- Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, 02115, USA
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Jeffrey D Dvorin
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, 02115, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
12
|
Rashpa R, Klages N, Schvartz D, Pasquarello C, Brochet M. The Skp1-Cullin1-FBXO1 complex is a pleiotropic regulator required for the formation of gametes and motile forms in Plasmodium berghei. Nat Commun 2023; 14:1312. [PMID: 36898988 PMCID: PMC10006092 DOI: 10.1038/s41467-023-36999-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 02/27/2023] [Indexed: 03/12/2023] Open
Abstract
Malaria-causing parasites of the Plasmodium genus undergo multiple developmental phases in the human and the mosquito hosts, regulated by various post-translational modifications. While ubiquitination by multi-component E3 ligases is key to regulate a wide range of cellular processes in eukaryotes, little is known about its role in Plasmodium. Here we show that Plasmodium berghei expresses a conserved SKP1/Cullin1/FBXO1 (SCFFBXO1) complex showing tightly regulated expression and localisation across multiple developmental stages. It is key to cell division for nuclear segregation during schizogony and centrosome partitioning during microgametogenesis. It is additionally required for parasite-specific processes including gamete egress from the host erythrocyte, as well as integrity of the apical and the inner membrane complexes (IMC) in merozoite and ookinete, two structures essential for the dissemination of these motile stages. Ubiquitinomic surveys reveal a large set of proteins ubiquitinated in a FBXO1-dependent manner including proteins important for egress and IMC organisation. We additionally demonstrate an interplay between FBXO1-dependent ubiquitination and phosphorylation via calcium-dependent protein kinase 1. Altogether we show that Plasmodium SCFFBXO1 plays conserved roles in cell division and is also important for parasite-specific processes in the mammalian and mosquito hosts.
Collapse
Affiliation(s)
- Ravish Rashpa
- University of Geneva, Faculty of Medicine, Department of Microbiology and Molecular Medicine, Geneva, Switzerland.
| | - Natacha Klages
- University of Geneva, Faculty of Medicine, Department of Microbiology and Molecular Medicine, Geneva, Switzerland
| | - Domitille Schvartz
- University of Geneva, Faculty of Medicine, Proteomics Core Facility, Geneva, Switzerland
| | - Carla Pasquarello
- University of Geneva, Faculty of Medicine, Proteomics Core Facility, Geneva, Switzerland
| | - Mathieu Brochet
- University of Geneva, Faculty of Medicine, Department of Microbiology and Molecular Medicine, Geneva, Switzerland.
| |
Collapse
|
13
|
Anam Z, Kumari G, Mukherjee S, Rex DAB, Biswas S, Maurya P, Ravikumar S, Gupta N, Kushawaha AK, Sah RK, Chaurasiya A, Singhal J, Singh N, Kaushik S, Prasad TSK, Pati S, Ranganathan A, Singh S. Complementary crosstalk between palmitoylation and phosphorylation events in MTIP regulates its role during Plasmodium falciparum invasion. Front Cell Infect Microbiol 2022; 12:924424. [PMID: 36250062 PMCID: PMC9556994 DOI: 10.3389/fcimb.2022.924424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/10/2022] [Indexed: 11/19/2022] Open
Abstract
Post-translational modifications (PTMs) including phosphorylation and palmitoylation have emerged as crucial biomolecular events that govern many cellular processes including functioning of motility- and invasion-associated proteins during Plasmodium falciparum invasion. However, no study has ever focused on understanding the possibility of a crosstalk between these two molecular events and its direct impact on preinvasion- and invasion-associated protein–protein interaction (PPI) network-based molecular machinery. Here, we used an integrated in silico analysis to enrich two different catalogues of proteins: (i) the first group defines the cumulative pool of phosphorylated and palmitoylated proteins, and (ii) the second group represents a common set of proteins predicted to have both phosphorylation and palmitoylation. Subsequent PPI analysis identified an important protein cluster comprising myosin A tail interacting protein (MTIP) as one of the hub proteins of the glideosome motor complex in P. falciparum, predicted to have dual modification with the possibility of a crosstalk between the same. Our findings suggested that blocking palmitoylation led to reduced phosphorylation and blocking phosphorylation led to abrogated palmitoylation of MTIP. As a result of the crosstalk between these biomolecular events, MTIP’s interaction with myosin A was found to be abrogated. Next, the crosstalk between phosphorylation and palmitoylation was confirmed at a global proteome level by click chemistry and the phenotypic effect of this crosstalk was observed via synergistic inhibition in P. falciparum invasion using checkerboard assay and isobologram method. Overall, our findings revealed, for the first time, an interdependence between two PTM types, their possible crosstalk, and its direct impact on MTIP-mediated invasion via glideosome assembly protein myosin A in P. falciparum. These insights can be exploited for futuristic drug discovery platforms targeting parasite molecular machinery for developing novel antimalarial therapeutics.
Collapse
Affiliation(s)
- Zille Anam
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Geeta Kumari
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Soumyadeep Mukherjee
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, Uttar Pradesh, India
| | | | - Shreeja Biswas
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Preeti Maurya
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Susendaran Ravikumar
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, Uttar Pradesh, India
| | - Nutan Gupta
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | | | - Raj Kumar Sah
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Ayushi Chaurasiya
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Jhalak Singhal
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Niharika Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Shikha Kaushik
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - T. S. Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore, India
| | - Soumya Pati
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, Uttar Pradesh, India
- *Correspondence: Shailja Singh, ; Anand Ranganathan, ; Soumya Pati,
| | - Anand Ranganathan
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
- *Correspondence: Shailja Singh, ; Anand Ranganathan, ; Soumya Pati,
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
- *Correspondence: Shailja Singh, ; Anand Ranganathan, ; Soumya Pati,
| |
Collapse
|
14
|
Jaramillo Ponce JR, Kapps D, Paulus C, Chicher J, Frugier M. Discovery of two distinct aminoacyl-tRNA synthetase complexes anchored to the Plasmodium surface tRNA import protein. J Biol Chem 2022; 298:101987. [PMID: 35487244 PMCID: PMC9136112 DOI: 10.1016/j.jbc.2022.101987] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 11/21/2022] Open
Abstract
Aminoacyl-tRNA synthetases (aaRSs) attach amino acids to their cognate transfer RNAs. In eukaryotes, a subset of cytosolic aaRSs is organized into a multisynthetase complex (MSC), along with specialized scaffolding proteins referred to as aaRS-interacting multifunctional proteins (AIMPs). In Plasmodium, the causative agent of malaria, the tRNA import protein (tRip), is a membrane protein that participates in tRNA trafficking; we show that tRip also functions as an AIMP. We identified three aaRSs, the glutamyl-tRNA synthetase (ERS), glutaminyl-tRNA synthetase (QRS), and methionyl-tRNA synthetase (MRS), which were specifically coimmunoprecipitated with tRip in Plasmodium berghei blood stage parasites. All four proteins contain an N-terminal glutathione-S-transferase (GST)-like domain that was demonstrated to be involved in MSC assembly. In contrast to previous studies, further dissection of GST-like interactions identified two exclusive heterotrimeric complexes: the Q-complex (tRip-ERS-QRS) and the M-complex (tRip-ERS-MRS). Gel filtration and light scattering suggest a 2:2:2 stoichiometry for both complexes but with distinct biophysical properties and mutational analysis further revealed that the GST-like domains of QRS and MRS use different strategies to bind ERS. Taken together, our results demonstrate that neither the singular homodimerization of tRip nor its localization in the parasite plasma membrane prevents the formation of MSCs in Plasmodium. Besides, the extracellular localization of the tRNA-binding module of tRip is compensated by the presence of additional tRNA-binding modules fused to MRS and QRS, providing each MSC with two spatially distinct functions: aminoacylation of intraparasitic tRNAs and binding of extracellular tRNAs. This unique host-pathogen interaction is discussed.
Collapse
Affiliation(s)
- José R Jaramillo Ponce
- Université de Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR 9002, Strasbourg, France
| | - Delphine Kapps
- Université de Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR 9002, Strasbourg, France
| | - Caroline Paulus
- Université de Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR 9002, Strasbourg, France
| | - Johana Chicher
- Strasbourg-Esplanade Proteomics Facility, Université de Strasbourg, Strasbourg, France
| | - Magali Frugier
- Université de Strasbourg, CNRS, Architecture et Réactivité de l'ARN, UPR 9002, Strasbourg, France.
| |
Collapse
|
15
|
A Phosphoinositide-Binding Protein Acts in the Trafficking Pathway of Hemoglobin in the Malaria Parasite Plasmodium falciparum. mBio 2022; 13:e0323921. [PMID: 35038916 PMCID: PMC8764524 DOI: 10.1128/mbio.03239-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Phosphoinositide lipids play key roles in a variety of processes in eukaryotic cells, but our understanding of their functions in the malaria parasite Plasmodium falciparum is still very much limited. To gain a deeper comprehension of the roles of phosphoinositides in this important pathogen, we attempted gene inactivation for 24 putative effectors of phosphoinositide metabolism. Our results reveal that 79% of the candidates are refractory to genetic deletion and are therefore potentially essential for parasite growth. Inactivation of the gene coding for a Plasmodium-specific putative phosphoinositide-binding protein, which we named PfPX1, results in a severe growth defect. We show that PfPX1 likely binds phosphatidylinositol-3-phosphate and that it localizes to the membrane of the digestive vacuole of the parasite and to vesicles filled with host cell cytosol and labeled with endocytic markers. Critically, we provide evidence that it is important in the trafficking pathway of hemoglobin from the host erythrocyte to the digestive vacuole. Finally, inactivation of PfPX1 renders parasites resistant to artemisinin, the frontline antimalarial drug. Globally, the minimal redundancy in the putative phosphoinositide proteins uncovered in our work supports that targeting this pathway has potential for antimalarial drug development. Moreover, our identification of a phosphoinositide-binding protein critical for the trafficking of hemoglobin provides key insight into this essential process. IMPORTANCE Malaria represents an enormous burden for a significant proportion of humanity, and the lack of vaccines and problems with drug resistance to all antimalarials demonstrate the need to develop new therapeutics. Inhibitors of phosphoinositide metabolism are currently being developed as antimalarials but our understanding of this biological pathway is incomplete. The malaria parasite lives inside human red blood cells where it imports hemoglobin to cover some of its nutritional needs. In this work, we have identified a phosphoinositide-binding protein that is important for the transport of hemoglobin in the parasite. Inactivation of this protein decreases the ability of the parasite to proliferate. Our results have therefore identified a potential new target for antimalarial development.
Collapse
|
16
|
Ndegwa DN, Kundu P, Hostetler JB, Marin-Menendez A, Sanderson T, Mwikali K, Verzier LH, Coyle R, Adjalley S, Rayner JC. Using Plasmodium knowlesi as a model for screening Plasmodium vivax blood-stage malaria vaccine targets reveals new candidates. PLoS Pathog 2021; 17:e1008864. [PMID: 34197567 PMCID: PMC8279373 DOI: 10.1371/journal.ppat.1008864] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 07/14/2021] [Accepted: 06/01/2021] [Indexed: 11/18/2022] Open
Abstract
Plasmodium vivax is responsible for the majority of malaria cases outside Africa. Unlike P. falciparum, the P. vivax life-cycle includes a dormant liver stage, the hypnozoite, which can cause infection in the absence of mosquito transmission. An effective vaccine against P. vivax blood stages would limit symptoms and pathology from such recurrent infections, and therefore could play a critical role in the control of this species. Vaccine development in P. vivax, however, lags considerably behind P. falciparum, which has many identified targets with several having transitioned to Phase II testing. By contrast only one P. vivax blood-stage vaccine candidate based on the Duffy Binding Protein (PvDBP), has reached Phase Ia, in large part because the lack of a continuous in vitro culture system for P. vivax limits systematic screening of new candidates. We used the close phylogenetic relationship between P. vivax and P. knowlesi, for which an in vitro culture system in human erythrocytes exists, to test the scalability of systematic reverse vaccinology to identify and prioritise P. vivax blood-stage targets. A panel of P. vivax proteins predicted to function in erythrocyte invasion were expressed as full-length recombinant ectodomains in a mammalian expression system. Eight of these antigens were used to generate polyclonal antibodies, which were screened for their ability to recognize orthologous proteins in P. knowlesi. These antibodies were then tested for inhibition of growth and invasion of both wild type P. knowlesi and chimeric P. knowlesi lines modified using CRISPR/Cas9 to exchange P. knowlesi genes with their P. vivax orthologues. Candidates that induced antibodies that inhibited invasion to a similar level as PvDBP were identified, confirming the utility of P. knowlesi as a model for P. vivax vaccine development and prioritizing antigens for further follow up.
Collapse
Affiliation(s)
- Duncan N. Ndegwa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Department of Biological Sciences, University of Embu, Embu, Kenya
| | - Prasun Kundu
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge, United Kingdom
| | - Jessica B. Hostetler
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | | | - Theo Sanderson
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Kioko Mwikali
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Lisa H. Verzier
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Rachael Coyle
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Sophie Adjalley
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Julian C. Rayner
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge, United Kingdom
| |
Collapse
|
17
|
Wichers JS, Wunderlich J, Heincke D, Pazicky S, Strauss J, Schmitt M, Kimmel J, Wilcke L, Scharf S, von Thien H, Burda PC, Spielmann T, Löw C, Filarsky M, Bachmann A, Gilberger TW. Identification of novel inner membrane complex and apical annuli proteins of the malaria parasite Plasmodium falciparum. Cell Microbiol 2021; 23:e13341. [PMID: 33830607 DOI: 10.1111/cmi.13341] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/29/2021] [Accepted: 04/05/2021] [Indexed: 02/06/2023]
Abstract
The inner membrane complex (IMC) is a defining feature of apicomplexan parasites, which confers stability and shape to the cell, functions as a scaffolding compartment during the formation of daughter cells and plays an important role in motility and invasion during different life cycle stages of these single-celled organisms. To explore the IMC proteome of the malaria parasite Plasmodium falciparum we applied a proximity-dependent biotin identification (BioID)-based proteomics approach, using the established IMC marker protein Photosensitized INA-Labelled protein 1 (PhIL1) as bait in asexual blood-stage parasites. Subsequent mass spectrometry-based peptide identification revealed enrichment of 12 known IMC proteins and several uncharacterized candidate proteins. We validated nine of these previously uncharacterized proteins by endogenous GFP-tagging. Six of these represent new IMC proteins, while three proteins have a distinct apical localization that most likely represents structures described as apical annuli in Toxoplasma gondii. Additionally, various Kelch13 interacting candidates were identified, suggesting an association of the Kelch13 compartment and the IMC in schizont and merozoite stages. This work extends the number of validated IMC proteins in the malaria parasite and reveals for the first time the existence of apical annuli proteins in P. falciparum. Additionally, it provides evidence for a spatial association between the Kelch13 compartment and the IMC in late blood-stage parasites.
Collapse
Affiliation(s)
- Jan Stephan Wichers
- Centre for Structural Systems Biology, Hamburg, Germany.,Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,University of Hamburg, Hamburg, Germany
| | - Juliane Wunderlich
- Centre for Structural Systems Biology, Hamburg, Germany.,European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany
| | - Dorothee Heincke
- Centre for Structural Systems Biology, Hamburg, Germany.,Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,University of Hamburg, Hamburg, Germany
| | - Samuel Pazicky
- Centre for Structural Systems Biology, Hamburg, Germany.,European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany
| | - Jan Strauss
- Centre for Structural Systems Biology, Hamburg, Germany.,Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,University of Hamburg, Hamburg, Germany
| | - Marius Schmitt
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Jessica Kimmel
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Louisa Wilcke
- Centre for Structural Systems Biology, Hamburg, Germany.,Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,University of Hamburg, Hamburg, Germany
| | - Sarah Scharf
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Heidrun von Thien
- Centre for Structural Systems Biology, Hamburg, Germany.,Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,University of Hamburg, Hamburg, Germany
| | - Paul-Christian Burda
- Centre for Structural Systems Biology, Hamburg, Germany.,Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,University of Hamburg, Hamburg, Germany
| | - Tobias Spielmann
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Christian Löw
- Centre for Structural Systems Biology, Hamburg, Germany.,European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany
| | - Michael Filarsky
- Centre for Structural Systems Biology, Hamburg, Germany.,University of Hamburg, Hamburg, Germany
| | - Anna Bachmann
- Centre for Structural Systems Biology, Hamburg, Germany.,Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,University of Hamburg, Hamburg, Germany.,German Centre for Infection Research (DZIF), partner site Hamburg-Borstel-Lübeck-Riems, Braunschweig, Germany
| | - Tim W Gilberger
- Centre for Structural Systems Biology, Hamburg, Germany.,Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,University of Hamburg, Hamburg, Germany
| |
Collapse
|
18
|
Morano AA, Dvorin JD. The Ringleaders: Understanding the Apicomplexan Basal Complex Through Comparison to Established Contractile Ring Systems. Front Cell Infect Microbiol 2021; 11:656976. [PMID: 33954122 PMCID: PMC8089483 DOI: 10.3389/fcimb.2021.656976] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/19/2021] [Indexed: 12/02/2022] Open
Abstract
The actomyosin contractile ring is a key feature of eukaryotic cytokinesis, conserved across many eukaryotic kingdoms. Recent research into the cell biology of the divergent eukaryotic clade Apicomplexa has revealed a contractile ring structure required for asexual division in the medically relevant genera Toxoplasma and Plasmodium; however, the structure of the contractile ring, known as the basal complex in these parasites, remains poorly characterized and in the absence of a myosin II homolog, it is unclear how the force required of a cytokinetic contractile ring is generated. Here, we review the literature on the basal complex in Apicomplexans, summarizing what is known about its formation and function, and attempt to provide possible answers to this question and suggest new avenues of study by comparing the Apicomplexan basal complex to well-studied, established cytokinetic contractile rings and their mechanisms in organisms such as S. cerevisiae and D. melanogaster. We also compare the basal complex to structures formed during mitochondrial and plastid division and cytokinetic mechanisms of organisms beyond the Opisthokonts, considering Apicomplexan diversity and divergence.
Collapse
Affiliation(s)
- Alexander A Morano
- Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, United States.,Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States
| | - Jeffrey D Dvorin
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
19
|
Absalon S, Dvorin JD. Depletion of the mini-chromosome maintenance complex binding protein allows the progression of cytokinesis despite abnormal karyokinesis during the asexual development of Plasmodium falciparum. Cell Microbiol 2020; 23:e13284. [PMID: 33124706 DOI: 10.1111/cmi.13284] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 12/23/2022]
Abstract
The eukaryotic cell cycle is typically divided into distinct phases with cytokinesis immediately following mitosis. To ensure proper cell division, each phase is tightly coordinated via feedback controls named checkpoints. During its asexual replication cycle, the malaria parasite Plasmodium falciparum undergoes multiple asynchronous rounds of mitosis with segregation of uncondensed chromosomes followed by nuclear division with intact nuclear envelope. The multi-nucleated schizont is then subjected to a single round of cytokinesis that produces dozens of daughter cells called merozoites. To date, no cell cycle checkpoints have been identified that regulate the Plasmodium spp. mode of division. Here, we identify the Plasmodium homologue of the Mini-Chromosome Maintenance Complex Binding Protein (PfMCMBP), which co-purified with the Mini-Chromosome Maintenance (MCM) complex, a replicative helicase required for genomic DNA replication. By conditionally depleting PfMCMBP, we disrupt nuclear morphology and parasite proliferation without causing a block in DNA replication. By immunofluorescence microscopy, we show that PfMCMBP depletion promotes the formation of mitotic spindle microtubules with extensions to more than one DNA focus and abnormal centrin distribution. Strikingly, PfMCMBP-deficient parasites complete cytokinesis and form aneuploid merozoites with variable cellular and nuclear sizes. Our study demonstrates that the parasite lacks a robust checkpoint response to prevent cytokinesis following aberrant karyokinesis.
Collapse
Affiliation(s)
- Sabrina Absalon
- Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jeffrey D Dvorin
- Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Pazicky S, Dhamotharan K, Kaszuba K, Mertens HDT, Gilberger T, Svergun D, Kosinski J, Weininger U, Löw C. Structural role of essential light chains in the apicomplexan glideosome. Commun Biol 2020; 3:568. [PMID: 33051581 PMCID: PMC7555893 DOI: 10.1038/s42003-020-01283-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 09/11/2020] [Indexed: 01/18/2023] Open
Abstract
Gliding, a type of motility based on an actin-myosin motor, is specific to apicomplexan parasites. Myosin A binds two light chains which further interact with glideosome associated proteins and assemble into the glideosome. The role of individual glideosome proteins is unclear due to the lack of structures of larger glideosome assemblies. Here, we investigate the role of essential light chains (ELCs) in Toxoplasma gondii and Plasmodium falciparum and present their crystal structures as part of trimeric sub-complexes. We show that although ELCs bind a conserved MyoA sequence, P. falciparum ELC adopts a distinct structure in the free and MyoA-bound state. We suggest that ELCs enhance MyoA performance by inducing secondary structure in MyoA and thus stiffen its lever arm. Structural and biophysical analysis reveals that calcium binding has no influence on the structure of ELCs. Our work represents a further step towards understanding the mechanism of gliding in Apicomplexa.
Collapse
Affiliation(s)
- Samuel Pazicky
- Centre for Structural Systems Biology (CSSB), Notkestrasse 85, D-22607, Hamburg, Germany
- Molecular Biology Laboratory (EMBL), Hamburg Unit c/o Deutsches Elektronen Synchrotron (DESY), Notkestrasse 85, D-22607, Hamburg, Germany
| | - Karthikeyan Dhamotharan
- Centre for Structural Systems Biology (CSSB), Notkestrasse 85, D-22607, Hamburg, Germany
- Molecular Biology Laboratory (EMBL), Hamburg Unit c/o Deutsches Elektronen Synchrotron (DESY), Notkestrasse 85, D-22607, Hamburg, Germany
| | - Karol Kaszuba
- Centre for Structural Systems Biology (CSSB), Notkestrasse 85, D-22607, Hamburg, Germany
- Molecular Biology Laboratory (EMBL), Hamburg Unit c/o Deutsches Elektronen Synchrotron (DESY), Notkestrasse 85, D-22607, Hamburg, Germany
| | - Haydyn D T Mertens
- Molecular Biology Laboratory (EMBL), Hamburg Unit c/o Deutsches Elektronen Synchrotron (DESY), Notkestrasse 85, D-22607, Hamburg, Germany
| | - Tim Gilberger
- Centre for Structural Systems Biology (CSSB), Notkestrasse 85, D-22607, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Strasse 74, D-20359, Hamburg, Germany
- Department of Biology, University of Hamburg, Hamburg, Germany
| | - Dmitri Svergun
- Molecular Biology Laboratory (EMBL), Hamburg Unit c/o Deutsches Elektronen Synchrotron (DESY), Notkestrasse 85, D-22607, Hamburg, Germany
| | - Jan Kosinski
- Centre for Structural Systems Biology (CSSB), Notkestrasse 85, D-22607, Hamburg, Germany
- Molecular Biology Laboratory (EMBL), Hamburg Unit c/o Deutsches Elektronen Synchrotron (DESY), Notkestrasse 85, D-22607, Hamburg, Germany
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Ulrich Weininger
- Martin-Luther-University Halle-Wittenberg, Institute of Physics, Biophysics, D-06120, Halle (Saale), Germany
| | - Christian Löw
- Centre for Structural Systems Biology (CSSB), Notkestrasse 85, D-22607, Hamburg, Germany.
- Molecular Biology Laboratory (EMBL), Hamburg Unit c/o Deutsches Elektronen Synchrotron (DESY), Notkestrasse 85, D-22607, Hamburg, Germany.
| |
Collapse
|
21
|
Amlabu E, Ilani P, Opoku G, Nyarko PB, Quansah E, Thiam LG, Anim M, Ayivor-Djanie R, Akuh OA, Mensah-Brown H, Rayner JC, Awandare GA. Molecular Characterization and Immuno-Reactivity Patterns of a Novel Plasmodium falciparum Armadillo-Type Repeat Protein, PfATRP. Front Cell Infect Microbiol 2020; 10:114. [PMID: 32266165 PMCID: PMC7100384 DOI: 10.3389/fcimb.2020.00114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 03/02/2020] [Indexed: 01/30/2023] Open
Abstract
Nearly half of the genes in the Plasmodium falciparum genome have not yet been functionally investigated. We used homology-based structural modeling to identify multiple copies of Armadillo repeats within one uncharacterized gene expressed during the intraerythrocytic stages, PF3D7_0410600, subsequently referred to as P. falciparum Armadillo-Type Repeat Protein (PfATRP). Soluble recombinant PfATRP was expressed in a bacterial expression system, purified to apparent homogeneity and the identity of the recombinant PfATRP was confirmed by mass spectrometry. Affinity-purified α-PfATRP rabbit antibodies specifically recognized the recombinant protein. Immunofluorescence assays revealed that α-PfATRP rabbit antibodies reacted with P. falciparum schizonts. Anti-PfATRP antibody exhibited peripheral staining patterns around the merozoites. Given the localization of PfATRP in merozoites, we tested for an egress phenotype during schizont arrest assays and demonstrated that native PfATRP is inaccessible on the surface of merozoites in intact schizonts. Dual immunofluorescence assays with markers for the inner membrane complex (IMC) and microtubules suggest partial colocalization in both asexual and sexual stage parasites. Using the soluble recombinant PfATRP in a screen of plasma samples revealed that malaria-infected children have naturally acquired PfATRP-specific antibodies.
Collapse
Affiliation(s)
- Emmanuel Amlabu
- West African Center for Cell Biology of Infectious Pathogens, University of Ghana, Accra, Ghana
- Department of Biochemistry, Kogi State University, Anyigba, Nigeria
| | - Philip Ilani
- West African Center for Cell Biology of Infectious Pathogens, University of Ghana, Accra, Ghana
| | - Grace Opoku
- West African Center for Cell Biology of Infectious Pathogens, University of Ghana, Accra, Ghana
| | - Prince B. Nyarko
- West African Center for Cell Biology of Infectious Pathogens, University of Ghana, Accra, Ghana
| | - Evelyn Quansah
- West African Center for Cell Biology of Infectious Pathogens, University of Ghana, Accra, Ghana
| | - Laty G. Thiam
- West African Center for Cell Biology of Infectious Pathogens, University of Ghana, Accra, Ghana
| | - Manfred Anim
- West African Center for Cell Biology of Infectious Pathogens, University of Ghana, Accra, Ghana
| | - Reuben Ayivor-Djanie
- West African Center for Cell Biology of Infectious Pathogens, University of Ghana, Accra, Ghana
- Department of Biomedical Sciences, SBBS, University of Health and Allied Sciences, Ho, Ghana
| | - Ojo-ajogu Akuh
- West African Center for Cell Biology of Infectious Pathogens, University of Ghana, Accra, Ghana
| | - Henrietta Mensah-Brown
- West African Center for Cell Biology of Infectious Pathogens, University of Ghana, Accra, Ghana
| | - Julian C. Rayner
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Gordon A. Awandare
- West African Center for Cell Biology of Infectious Pathogens, University of Ghana, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| |
Collapse
|
22
|
Ebrahimzadeh Z, Mukherjee A, Crochetière MÈ, Sergerie A, Amiar S, Thompson LA, Gagnon D, Gaumond D, Stahelin RV, Dacks JB, Richard D. A pan-apicomplexan phosphoinositide-binding protein acts in malarial microneme exocytosis. EMBO Rep 2019; 20:e47102. [PMID: 31097469 PMCID: PMC6549027 DOI: 10.15252/embr.201847102] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 03/28/2019] [Accepted: 04/12/2019] [Indexed: 11/09/2022] Open
Abstract
Invasion of human red blood cells by the malaria parasite Plasmodium falciparum is an essential step in the development of the disease. Consequently, the molecular players involved in host cell invasion represent important targets for inhibitor design and vaccine development. The process of merozoite invasion is a succession of steps underlined by the sequential secretion of the organelles of the apical complex. However, little is known with regard to how their contents are exocytosed. Here, we identify a phosphoinositide-binding protein conserved in apicomplexan parasites and show that it is important for the attachment and subsequent invasion of the erythrocyte by the merozoite. Critically, removing the protein from its site of action by knock sideways preferentially prevents the secretion of certain types of micronemes. Our results therefore provide evidence for a role of phosphoinositide lipids in the malaria invasion process and provide further insight into the secretion of microneme organelle populations, which is potentially applicable to diverse apicomplexan parasites.
Collapse
Affiliation(s)
- Zeinab Ebrahimzadeh
- Centre de Recherche en Infectiologie, CRCHU de Québec-Université Laval, Québec, QC, Canada
| | - Angana Mukherjee
- Centre de Recherche en Infectiologie, CRCHU de Québec-Université Laval, Québec, QC, Canada
| | - Marie-Ève Crochetière
- Centre de Recherche en Infectiologie, CRCHU de Québec-Université Laval, Québec, QC, Canada
| | - Audrey Sergerie
- Centre de Recherche en Infectiologie, CRCHU de Québec-Université Laval, Québec, QC, Canada
| | - Souad Amiar
- Department of Medicinal Chemistry and Molecular Pharmacology and the Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, USA
| | - L Alexa Thompson
- Division of Infectious Disease, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Dominic Gagnon
- Centre de Recherche en Infectiologie, CRCHU de Québec-Université Laval, Québec, QC, Canada
| | - David Gaumond
- Centre de Recherche en Infectiologie, CRCHU de Québec-Université Laval, Québec, QC, Canada
| | - Robert V Stahelin
- Department of Medicinal Chemistry and Molecular Pharmacology and the Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, USA
| | - Joel B Dacks
- Division of Infectious Disease, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Dave Richard
- Centre de Recherche en Infectiologie, CRCHU de Québec-Université Laval, Québec, QC, Canada
| |
Collapse
|
23
|
Bwire GM, Majigo M, Makalla R, Nkinda L, Mawazo A, Mizinduko M, Makani J. Immunoglobulin G responses against falciparum malaria specific antigens are higher in children with homozygous sickle cell trait than those with normal hemoglobin. BMC Immunol 2019; 20:12. [PMID: 31029083 PMCID: PMC6486967 DOI: 10.1186/s12865-019-0294-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/12/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND High Immunoglobulin G (IgG) response to Plasmodium falciparum antigens is associated with partial malaria protection in sickle hemoglobin (HbS) children. However, this response has been more studied in children with heterozygous sickle cell trait (HbAS) but little explored in those with homozygous sickle cell trait (HbSS). The current study was conducted to determine the IgG responses against specific Plasmodium falciparum antigens in children with homozygous sickle cell trait (HbSS) by comparing to those with normal hemoglobin (HbAA). METHODS A cross sectional study was conducted between April and July 2018 in Dar es Salaam tertiary hospitals. Parents were consented for their child to give about 5 ml of venous blood. IgG concentration from the blood plasma of 220 children (110 HbAA vs. 110 HbSS) were determined using indirect Enzyme Linked Immunosorbent Assay (ELISA). Then IgG medians were compared between the groups with prism 5 software (GraphPad) using Mann Whitney U test. Where the differences in age, hemoglobin levels and body weight between the groups was analyzed using independent sample t test. Multiple linear regressions were used to control cofounding variables such as body weight, age and hemoglobin level using statistical package for social sciences software (SPSS version 23). P value <0.05 was considered statistically significant. RESULTS The median IgG concentration to PfEBA-175, Pfg27, yPfs28C antigens were HbSS; 20.7 ng/ml (IQR; 18.1-25.6) vs. HbAA; 2.3 ng/ml (IQR; 1.21-3.04), HbSS; 2.76 ng/ml (IQR: 2.08-5.69) vs. HbAA; 1.36 ng/ml (IQR: 1.28-1.76), and HbSS; 26,592 ng/ml (IQR: 10817-41,462) vs. HbAA; 14,164 ng/ml (IQR; 3069-24,302) respectively (p < 0.0001 for all IgG). In both groups; age, body weight and hemoglobin level had no impact on the levels of IgG responses to Plasmodium falciparum antigens except for HbAA group which showed a significant increase in IgG against Pfg27 by 0.004 ng/ml with 1 g/dl increase in Hb level (p = 0.028). CONCLUSIONS This study found significant higher levels of specific Plasmodium falciparum IgG responses in children with homozygous sickle cell trait than those with normal hemoglobin.
Collapse
Affiliation(s)
- George Msema Bwire
- Department of Pharmaceutical Microbiology, School of Pharmacy, Muhimbili University of Health and Allied Sciences, Box 65001, Dar es Salaam, Tanzania. .,Department of Microbiology and Immunology, School of Medicine, Muhimbili University of Health and Allied Sciences, Box 65001, Dar es Salaam, Tanzania.
| | - Mtebe Majigo
- Department of Microbiology and Immunology, School of Medicine, Muhimbili University of Health and Allied Sciences, Box 65001, Dar es Salaam, Tanzania
| | - Robert Makalla
- Department of Microbiology and Immunology, School of Medicine, Muhimbili University of Health and Allied Sciences, Box 65001, Dar es Salaam, Tanzania.,Ministry of Health, Community Development, Gender, Elderly and Children, Box 143, Babati, Manyara, Tanzania
| | - Lillian Nkinda
- Department of Microbiology and Immunology, School of Medicine, Muhimbili University of Health and Allied Sciences, Box 65001, Dar es Salaam, Tanzania
| | - Akili Mawazo
- Department of Microbiology and Immunology, School of Medicine, Muhimbili University of Health and Allied Sciences, Box 65001, Dar es Salaam, Tanzania
| | - Mucho Mizinduko
- Department of Epidemiology and Biostatistics, School of Public Health, Muhimbili University of Health and Allied Sciences, Box 65001, Dar es Salaam, Tanzania
| | - Julie Makani
- Department of Hematology and Blood Transfusion, School of Medicine, Muhimbili University of Health and Allied Sciences, Box 65001, Dar es Salaam, Tanzania
| |
Collapse
|
24
|
Identification and molecular characterization of a novel Babesia orientalis thrombospondin-related anonymous protein (BoTRAP1). Parasit Vectors 2018; 11:667. [PMID: 30587207 PMCID: PMC6307320 DOI: 10.1186/s13071-018-3245-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/29/2018] [Indexed: 11/18/2022] Open
Abstract
Background The thrombospondin-related anonymous protein (TRAP) family, a kind of transmembrane protein, is widely distributed with a conserved feature of structure in all apicomplexan parasites and plays a crucial role in the gliding motility and survival of parasites. Methods The Babesia orientalis TRAP1 gene (BoTRAP1) was truncated and cloned into a pET-42b expression vector and expressed as a GST-tag fusion protein with a TEV protease site. Rabbit anti-rBoTRAP1 antibody was produced and purified using a protein A chromatography column. Western blot analysis was performed to identify the native protein of BoTRAP1 and differentiate B. orientalis-infected positive from negative serum samples. The localization of BoTRAP1 on merozoites was identified by the indirect florescent antibody test (IFAT). Results The partial sequence of the TRAP1 gene was cloned from B. orientalis cDNA and identified to contain a von Willebrand factor A (vWFA) region and a thrombospondin type-1 (TSP-1) domain; it had a length of 762 bp, encoding a polypeptide of 254 amino acid residues with a predicted size of 28.2 kDa. The partial sequence was cloned into a pET-42b expression vector and expressed in E. coli as a GST fusion protein. Western blot indicated that rBoTRAP1 has a high immunogenicity and can differentiate B. orientalis-infected positive and negative serum samples collected from water buffaloes. IFAT showed that BoTRAP1 is mainly localized on the apical end of intracellular parasites by using polyclonal antibodies (PcAb) against rBoTRAP1. Meanwhile, the PcAb test also identified the native BoTRAP1 as a ~65 kDa band from B. orientalis lysates. The predicted 3D structure of BoTRAP1 contains a metalion-dependent adhesion site (MIDAS), which could be important for interaction with ligand on the surface of the host cells. Conclusions Like all known protozoa, B. orientalis has a TRAP family, comprising TRAP1, TRAP2, TRAP3 and TRAP4. The newly identified and characterized BoTRAP1 may play a key role in the invasion of B. orientalis into water buffalo erythrocytes.
Collapse
|
25
|
Plasmodium falciparum Cyclic GMP-Dependent Protein Kinase Interacts with a Subunit of the Parasite Proteasome. Infect Immun 2018; 87:IAI.00523-18. [PMID: 30323024 DOI: 10.1128/iai.00523-18] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/29/2018] [Indexed: 12/20/2022] Open
Abstract
Malaria is caused by the protozoan parasite Plasmodium, which undergoes a complex life cycle in a human host and a mosquito vector. The parasite's cyclic GMP (cGMP)-dependent protein kinase (PKG) is essential at multiple steps of the life cycle. Phosphoproteomic studies in Plasmodium falciparum erythrocytic stages and Plasmodium berghei ookinetes have identified proteolysis as a major biological pathway dependent on PKG activity. To further understand PKG's mechanism of action, we screened a yeast two-hybrid library for P. falciparum proteins that interact with P. falciparum PKG (PfPKG) and tested peptide libraries to identify its phosphorylation site preferences. Our data suggest that PfPKG has a distinct phosphorylation site and that PfPKG directly phosphorylates parasite RPT1, one of six AAA+ ATPases present in the 19S regulatory particle of the proteasome. PfPKG and RPT1 interact in vitro, and the interacting fragment of RPT1 carries a PfPKG consensus phosphorylation site; a peptide carrying this consensus site competes with the RPT1 fragment for binding to PfPKG and is efficiently phosphorylated by PfPKG. These data suggest that PfPKG's phosphorylation of RPT1 could contribute to its regulation of parasite proteolysis. We demonstrate that proteolysis plays an important role in a biological process known to require Plasmodium PKG: invasion by sporozoites of hepatocytes. A small-molecule inhibitor of proteasomal activity blocks sporozoite invasion in an additive manner when combined with a Plasmodium PKG-specific inhibitor. Mining the previously described parasite PKG-dependent phosphoproteomes using the consensus phosphorylation motif identified additional proteins that are likely to be direct substrates of the enzyme.
Collapse
|
26
|
Deligianni E, Silmon de Monerri NC, McMillan PJ, Bertuccini L, Superti F, Manola M, Spanos L, Louis C, Blackman MJ, Tilley L, Siden-Kiamos I. Essential role of Plasmodium perforin-like protein 4 in ookinete midgut passage. PLoS One 2018; 13:e0201651. [PMID: 30102727 PMCID: PMC6089593 DOI: 10.1371/journal.pone.0201651] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/19/2018] [Indexed: 01/22/2023] Open
Abstract
Pore forming proteins such as those belonging to the membrane attack/perforin (MACPF) family have important functions in many organisms. Of the five MACPF proteins found in Plasmodium parasites, three have functions in cell passage and one in host cell egress. Here we report an analysis of the perforin-like protein 4, PPLP4, in the rodent parasite Plasmodium berghei. We found that the protein is expressed only in the ookinete, the invasive stage of the parasite formed in the mosquito midgut. Transcriptional analysis revealed that expression of the pplp4 gene commences during ookinete development. The protein was detected in retorts and mature ookinetes. Using two antibodies, the protein was found localized in a dotted pattern, and 3-D SIM super-resolution microcopy revealed the protein in the periphery of the cell. Analysis of a C-terminal mCherry fusion of the protein however showed mainly cytoplasmic label. A pplp4 null mutant formed motile ookinetes, but these were unable to invade and traverse the midgut epithelium resulting in severely impaired oocyst formation and no transmission to naïve mice. However, when in vitro cultured ookinetes were injected into the thorax of the mosquito, thus by-passing midgut passage, sporozoites were formed and the mutant parasites were able to infect naïve mice. Taken together, our data show that PPLP4 is required only for ookinete invasion of the mosquito midgut. Thus PPLP4 has a similar role to the previously studied PPLP3 and PPLP5, raising the question why three proteins with MACPF domains are needed for invasion by the ookinete of the mosquito midgut epithelium.
Collapse
Affiliation(s)
- Elena Deligianni
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology—Hellas, Heraklion, Greece
- * E-mail:
| | | | - Paul J. McMillan
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, VIC, Australia
- ARC Centre of Excellence for Coherent X-ray Science, The University of Melbourne, Melbourne, VIC, Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, Australia
- Biological Optical Microcopy Platform, The University of Melbourne, Melbourne, VIC, Australia
| | - Lucia Bertuccini
- National Centre for Innovative Technologies in Public Health, National Institute of Health, Rome, Italy
| | - Fabiana Superti
- National Centre for Innovative Technologies in Public Health, National Institute of Health, Rome, Italy
| | - Maria Manola
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology—Hellas, Heraklion, Greece
| | - Lefteris Spanos
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology—Hellas, Heraklion, Greece
| | - Christos Louis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology—Hellas, Heraklion, Greece
| | - Michael J. Blackman
- The Francis Crick Institute, London, United Kingdom
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Leann Tilley
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, VIC, Australia
- ARC Centre of Excellence for Coherent X-ray Science, The University of Melbourne, Melbourne, VIC, Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Inga Siden-Kiamos
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology—Hellas, Heraklion, Greece
| |
Collapse
|
27
|
In vitro interaction between Plasmodium falciparum myosin B (PfMyoB) and myosin A tail interacting protein (MTIP). Parasitol Res 2018; 117:3437-3446. [PMID: 30094538 DOI: 10.1007/s00436-018-6039-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 08/02/2018] [Indexed: 10/28/2022]
Abstract
Apicomplexan parasites, including Plasmodium falciparum, are obligate intracellular organisms that utilize a strategy termed "gliding" to move and invade host cells, causing disease. Gliding is carried out by a protein complex known as the glideosome, which includes an actin-myosin motor. To date, six myosins have been identified in P. falciparum (PfMyoA, B, C, D, E, and F), but only the role of PfMyoA, the myosin of the glideosome that is involved in the process of red blood cell and mosquito cell invasion, has been established. Based on previous observations, we speculated that PfMyoA and PfMyoB may have similar or redundant functions. To test this hypothesis, we searched for in vitro interactions between PfMyoB and MTIP (myosin A tail interacting protein), the myosin light chain of PfMyoA. A set of differentially tagged PfMyoA, PfMyoB, and MTIP recombinant proteins was employed to specifically and simultaneously detect each myosin in competition assays and inhibition assays using specific peptides. MTIP potentially acts as the light chain of PfMyoB.
Collapse
|
28
|
Abstract
Apicomplexa are obligate intracellular parasites that actively invade, replicate within, and egress from host cells. The parasite actinomyosin-based molecular motor complex (often referred to as the glideosome) is considered an important mediator of parasite motility and virulence. Mature intracellular parasites often become motile just prior to egress from their host cells, and in some genera, this motility is important for successful egress as well as for subsequent invasion of new host cells. To determine whether actinomyosin-based motility is important in the red blood cell egress and invasion activities of the malaria parasite, we have used a conditional genetic approach to delete GAP45, a primary component of the glideosome, in asexual blood stages of Plasmodium falciparum Our results confirm the essential nature of GAP45 for invasion but show that P. falciparum does not require a functional motor complex to undergo egress from the red blood cell. Malarial egress therefore differs fundamentally from induced egress in the related apicomplexan Toxoplasma gondiiIMPORTANCE Clinical malaria results from cycles of replication of single-celled parasites of the genus Plasmodium in red blood cells. Intracellular parasite replication is followed by a highly regulated, protease-dependent process called egress, in which rupture of the bounding membranes allows explosive release of daughter merozoites which rapidly invade fresh red cells. A parasite actinomyosin-based molecular motor (the glideosome) has been proposed to provide the mechanical force to drive invasion. Studies of the related parasite Toxoplasma gondii have shown that induced egress requires parasite motility, mediated by a functional glideosome. However, whether the glideosome has a similar essential role in egress of malaria merozoites from red blood cells is unknown. Here, we show that although a functional glideosome is required for red blood cell invasion by Plasmodium falciparum merozoites, it is not required for egress. These findings place further emphasis on the key role of the protease cascade in malarial egress.
Collapse
|
29
|
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: 2.7] [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
|
30
|
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: 0.8] [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
|
31
|
Calcium-Dependent Protein Kinase 5 Is Required for Release of Egress-Specific Organelles in Plasmodium falciparum. mBio 2018; 9:mBio.00130-18. [PMID: 29487234 PMCID: PMC5829822 DOI: 10.1128/mbio.00130-18] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The human malaria parasite Plasmodium falciparum requires efficient egress out of an infected red blood cell for pathogenesis. This egress event is highly coordinated and is mediated by several signaling proteins, including the plant-like Pfalciparum calcium-dependent protein kinase 5 (PfCDPK5). Knockdown of PfCDPK5 results in an egress block where parasites are trapped inside their host cells. The mechanism of this PfCDPK5-dependent block, however, remains unknown. Here, we show that PfCDPK5 colocalizes with a specialized set of parasite organelles known as micronemes and is required for their discharge, implicating failure of this step as the cause of the egress defect in PfCDPK5-deficient parasites. Furthermore, we show that PfCDPK5 cooperates with the Pfalciparum cGMP-dependent kinase (PfPKG) to fully activate the protease cascade critical for parasite egress. The PfCDPK5-dependent arrest can be overcome by hyperactivation of PfPKG or by physical disruption of the arrested parasite, and we show that both treatments facilitate the release of the micronemes required for egress. Our results define the molecular mechanism of PfCDPK5 function and elucidate the complex signaling pathway of parasite egress.IMPORTANCE The signs and symptoms of clinical malaria result from the replication of parasites in human blood. Efficient egress of the malaria parasite Plasmodium falciparum out of an infected red blood cell is critical for pathogenesis. The Pfalciparum calcium-dependent protein kinase 5 (PfCDPK5) is essential for parasite egress. Following PfCDPK5 knockdown, parasites remain trapped inside their host cell and do not egress, but the mechanism for this block remains unknown. We show that PfCDPK5 colocalizes with parasite organelles known as micronemes. We demonstrate that PfCDPK5 is critical for the discharge of these micronemes and that failure of this step is the molecular mechanism of the parasite egress arrest. We also show that hyperactivation of the cGMP-dependent kinase PKG can overcome this arrest. Our data suggest that small molecules that inhibit the egress signaling pathway could be effective antimalarial therapeutics.
Collapse
|
32
|
Tannous S, Ghanem E. A bite to fight: front-line innate immune defenses against malaria parasites. Pathog Glob Health 2018; 112:1-12. [PMID: 29376476 DOI: 10.1080/20477724.2018.1429847] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Malaria infection caused by Plasmodium parasites remains a major health burden worldwide especially in the tropics and subtropics. Plasmodium exhibits a complex life cycle whereby it undergoes a series of developmental stages in the Anopheles mosquito vector and the vertebrate human host. Malaria severity is mainly attributed to the genetic complexity of the parasite which is reflected in the sophisticated mechanisms of invasion and evasion that allow it to overcome the immune responses of both its invertebrate and vertebrate hosts. In this review, we aim to provide an updated, clear and concise summary of the literature focusing on the interactions of the vertebrate innate immune system with Plasmodium parasites, namely sporozoites, merozoites, and trophozoites. The roles of innate immune factors, both humoral and cellular, in anti-Plasmodium defense are described with particular emphasis on the contribution of key innate players including neutrophils, macrophages, and natural killer cells to the clearance of liver and blood stage parasites. A comprehensive understanding of the innate immune responses to malaria parasites remains an important goal that would dramatically help improve the design of original treatment strategies and vaccines, both of which are urgently needed to relieve the burden of malaria especially in endemic countries.
Collapse
Affiliation(s)
- Stephanie Tannous
- a Faculty of Natural and Applied Sciences, Department of Sciences , Notre Dame University , Louaize , Lebanon
| | - Esther Ghanem
- a Faculty of Natural and Applied Sciences, Department of Sciences , Notre Dame University , Louaize , Lebanon
| |
Collapse
|
33
|
Nyagwange J, Tijhaar E, Ternette N, Mobegi F, Tretina K, Silva JC, Pelle R, Nene V. Characterization of the Theileria parva sporozoite proteome. Int J Parasitol 2017; 48:265-273. [PMID: 29258832 PMCID: PMC5854367 DOI: 10.1016/j.ijpara.2017.09.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/16/2017] [Accepted: 09/21/2017] [Indexed: 12/29/2022]
Abstract
2007 Theileria parva proteins expressed in the sporozoite were identified. Proteins include known T. parva antigens targeted by antibodies and cytotoxic T cells. Proteins predicted to be orthologs of Plasmodium falciparum sporozoite surface molecules were identified. Proteins predicted to be orthologs of P. falciparum invasion organelle proteins were identified. Proteins that may contribute to the phenomenon of bovine lymphocyte transformation were identified.
East Coast fever is a lymphoproliferative disease caused by the tick-borne protozoan parasite Theileria parva. The sporozoite stage of this parasite, harboured and released from the salivary glands of the tick Rhipicephalus appendiculatus during feeding, invades and establishes infection in bovine lymphocytes. Blocking this initial stage of invasion presents a promising vaccine strategy for control of East Coast fever and can in part be achieved by targeting the major sporozoite surface protein p67. To support research on the biology of T. parva and the identification of additional candidate vaccine antigens, we report on the sporozoite proteome as defined by LC–MS/MS analysis. In total, 4780 proteins were identified in an enriched preparation of sporozoites. Of these, 2007 were identified as T. parva proteins, representing close to 50% of the total predicted parasite proteome. The remaining 2773 proteins were derived from the tick vector. The identified sporozoite proteins include a set of known T. parva antigens targeted by antibodies and cytotoxic T cells from cattle that are immune to East Coast fever. We also identified proteins predicted to be orthologs of Plasmodium falciparum sporozoite surface molecules and invasion organelle proteins, and proteins that may contribute to the phenomenon of bovine lymphocyte transformation. Overall, these data establish a protein expression profile of T. parva sporozoites as an important starting point for further study of a parasitic species which has considerable agricultural impact.
Collapse
Affiliation(s)
- James Nyagwange
- International Livestock Research Institute, P.O. Box 30709, Nairobi, Kenya; Cell Biology and Immunology Group, Wageningen University, The Netherlands
| | - Edwin Tijhaar
- Cell Biology and Immunology Group, Wageningen University, The Netherlands
| | - Nicola Ternette
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, UK
| | - Fredrick Mobegi
- Department of Infection and Immunity, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, South Australia, Australia
| | - Kyle Tretina
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joana C Silva
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Roger Pelle
- International Livestock Research Institute, P.O. Box 30709, Nairobi, Kenya
| | - Vishvanath Nene
- International Livestock Research Institute, P.O. Box 30709, Nairobi, Kenya.
| |
Collapse
|
34
|
Mueller C, Graindorge A, Soldati-Favre D. Functions of myosin motors tailored for parasitism. Curr Opin Microbiol 2017; 40:113-122. [DOI: 10.1016/j.mib.2017.11.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 11/02/2017] [Accepted: 11/02/2017] [Indexed: 01/01/2023]
|
35
|
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: 1.7] [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
|
36
|
The Malaria Parasite Cyclin H Homolog PfCyc1 Is Required for Efficient Cytokinesis in Blood-Stage Plasmodium falciparum. mBio 2017; 8:mBio.00605-17. [PMID: 28611247 PMCID: PMC5472185 DOI: 10.1128/mbio.00605-17] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
All well-studied eukaryotic cell cycles are driven by cyclins, which activate cyclin-dependent kinases (CDKs), and these protein kinase complexes are viable drug targets. The regulatory control of the Plasmodium falciparum cell division cycle remains poorly understood, and the roles of the various CDKs and cyclins remain unclear. The P. falciparum genome contains multiple CDKs, but surprisingly, it does not contain any sequence-identifiable G1-, S-, or M-phase cyclins. We demonstrate that P. falciparum Cyc1 (PfCyc1) complements a G1 cyclin-depleted Saccharomyces cerevisiae strain and confirm that other identified malaria parasite cyclins do not complement this strain. PfCyc1, which has the highest sequence similarity to the conserved cyclin H, cannot complement a temperature-sensitive yeast cyclin H mutant. Coimmunoprecipitation of PfCyc1 from P. falciparum parasites identifies PfMAT1 and PfMRK as specific interaction partners and does not identify PfPK5 or other CDKs. We then generate an endogenous conditional allele of PfCyc1 in blood-stage P. falciparum using a destabilization domain (DD) approach and find that PfCyc1 is essential for blood-stage proliferation. PfCyc1 knockdown does not impede nuclear division, but it prevents proper cytokinesis. Thus, we demonstrate that PfCyc1 has a functional divergence from bioinformatic predictions, suggesting that the malaria parasite cell division cycle has evolved to use evolutionarily conserved proteins in functionally novel ways. Human infection by the eukaryotic parasite Plasmodium falciparum causes malaria. Most well-studied eukaryotic cell cycles are driven by cyclins, which activate cyclin-dependent kinases (CDKs) to promote essential cell division processes. Remarkably, there are no identifiable cyclins that are predicted to control the cell cycle in the malaria parasite genome. Thus, our knowledge regarding the basic mechanisms of the malaria parasite cell cycle remains unsatisfactory. We demonstrate that P. falciparum Cyc1 (PfCyc1), a transcriptional cyclin homolog, complements a cell cycle cyclin-deficient yeast strain but not a transcriptional cyclin-deficient strain. We show that PfCyc1 forms a complex in the parasite with PfMRK and the P. falciparum MAT1 homolog. PfCyc1 is essential and nonredundant in blood-stage P. falciparum. PfCyc1 knockdown causes a stage-specific arrest after nuclear division, demonstrating morphologically aberrant cytokinesis. This work demonstrates a conserved PfCyc1/PfMAT1/PfMRK complex in malaria and suggests that it functions as a schizont stage-specific regulator of the P. falciparum life cycle.
Collapse
|
37
|
Tardieux I, Baum J. Reassessing the mechanics of parasite motility and host-cell invasion. J Cell Biol 2017; 214:507-15. [PMID: 27573462 PMCID: PMC5004448 DOI: 10.1083/jcb.201605100] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 08/09/2016] [Indexed: 12/20/2022] Open
Abstract
The capacity to migrate is fundamental to multicellular and single-celled life. Apicomplexan parasites, an ancient protozoan clade that includes malaria parasites (Plasmodium) and Toxoplasma, achieve remarkable speeds of directional cell movement. This rapidity is achieved via a divergent actomyosin motor system, housed within a narrow compartment that lies underneath the length of the parasite plasma membrane. How this motor functions at a mechanistic level during motility and host cell invasion is a matter of debate. Here, we integrate old and new insights toward refining the current model for the function of this motor with the aim of revitalizing interest in the mechanics of how these deadly pathogens move.
Collapse
Affiliation(s)
- Isabelle Tardieux
- Institute of Advanced BioSciences, Institut National de la Santé et de la Recherche Médicale U1209, Centre National de la Recherche Scientifique UMR 5309, Université Grenoble Alpes, 38000, Grenoble, France
| | - Jake Baum
- Department of Life Sciences, Imperial College London, London SW7 2AZ, England, UK
| |
Collapse
|
38
|
P113 is a merozoite surface protein that binds the N terminus of Plasmodium falciparum RH5. Nat Commun 2017; 8:14333. [PMID: 28186186 PMCID: PMC5309799 DOI: 10.1038/ncomms14333] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 12/16/2016] [Indexed: 01/12/2023] Open
Abstract
Invasion of erythrocytes by Plasmodium falciparum merozoites is necessary for malaria pathogenesis and is therefore a primary target for vaccine development. RH5 is a leading subunit vaccine candidate because anti-RH5 antibodies inhibit parasite growth and the interaction with its erythrocyte receptor basigin is essential for invasion. RH5 is secreted, complexes with other parasite proteins including CyRPA and RIPR, and contains a conserved N-terminal region (RH5Nt) of unknown function that is cleaved from the native protein. Here, we identify P113 as a merozoite surface protein that directly interacts with RH5Nt. Using recombinant proteins and a sensitive protein interaction assay, we establish the binding interdependencies of all the other known RH5 complex components and conclude that the RH5Nt-P113 interaction provides a releasable mechanism for anchoring RH5 to the merozoite surface. We exploit these findings to design a chemically synthesized peptide corresponding to RH5Nt, which could contribute to a cost-effective malaria vaccine. The secreted Plasmodium falciparum protein RH5 is essential for invasion of erythrocytes and is a promising vaccine candidate. Here, Galaway et al. show that the N-terminal region of RH5 binds the GPI-anchored merozoite surface protein P113 and can elicit invasion-blocking antibodies.
Collapse
|
39
|
Receptor for Activated C-Kinase 1 (PfRACK1) is required for Plasmodium falciparum intra-erythrocytic proliferation. Mol Biochem Parasitol 2016; 211:62-66. [PMID: 27732881 DOI: 10.1016/j.molbiopara.2016.10.002] [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: 08/17/2016] [Revised: 10/05/2016] [Accepted: 10/08/2016] [Indexed: 11/24/2022]
Abstract
Emerging resistance to current anti-malarials necessitates a more detailed understanding of the biological processes of Plasmodium falciparum proliferation, thus allowing identification of new drug targets. The well-conserved protein Receptor for Activated C-Kinase 1 (RACK1) was originally identified in mammalian cells as an anchoring protein for protein kinase C (PKC) and has since been shown to be important for cell migration, cytokinesis, transcription, epigenetics, and protein translation. The P. falciparum ortholog, PfRACK1, is expressed in blood stages of the parasite and is diffusely localized in the parasite cytoplasm. Using a destabilizing domain to allow inducible knockdown of the endogenous protein level, we evaluated the requirement for PfRACK1 during blood-stage replication. Following destabilization, the parasites demonstrate a nearly complete growth arrest at the trophozoite stage. The essential nature of PfRACK1 suggests that the protein itself or the pathways regulated by the protein are potential targets for novel anti-malarial therapeutics.
Collapse
|
40
|
Production of recombinant proteins from Plasmodium falciparum in Escherichia coli. BIOMEDICA 2016; 36:97-108. [PMID: 27622630 DOI: 10.7705/biomedica.v36i3.3011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 11/16/2015] [Indexed: 01/30/2023]
Abstract
INTRODUCTION The production of recombinant proteins is essential for the characterization and functional study of proteins from Plasmodium falciparum. However, the proteins of P. falciparum are among the most challenging to express, and when expression is achieved, the recombinant proteins usually fold incorrectly and lead to the formation of inclusion bodies. OBJECTIVE To obtain and purify four recombinant proteins and to use them as antigens to produce polyclonal antibodies. The production efficiency and solubility were evaluated as the proteins were expressed in two genetically modified strains of Escherichia coli to favor the production of heterologous proteins (BL21-CodonPlus (DE3)-RIL and BL21-pG-KJE8). MATERIALS AND METHODS The four recombinant P. falciparum proteins corresponding to partial sequences of PfMyoA (Myosin A) and PfGAP50 (gliding associated protein 50), and the complete sequences of PfMTIP (myosin tail interacting protein) and PfGAP45 (gliding associated protein 45), were produced as glutathione S-transferase-fusion proteins, purified and used for immunizing mice. RESULTS The protein expression was much more efficient in BL21-CodonPlus, the strain that contains tRNAs that are rare in wild-type E. coli, compared to the expression in BL21-pG-KJE8. In spite of the fact that BL21-pG-KJE8 overexpresses chaperones, this strain did not minimize the formation of inclusion bodies. CONCLUSION The use of genetically modified strains of E. coli was essential to achieve high expression levels of the four evaluated P. falciparum proteins and lead to improved solubility of two of them. The approach used here allowed us to obtain and purify four P. falciparum proteins in enough quantity to produce polyclonal antibodies in mice, and a fair amount of two pure and soluble recombinant proteins for future assays.
Collapse
|
41
|
Han JH, Li J, Wang B, Lee SK, Nyunt MH, Na S, Park JH, Han ET. Identification of Immunodominant B-cell Epitope Regions of Reticulocyte Binding Proteins in Plasmodium vivax by Protein Microarray Based Immunoscreening. THE KOREAN JOURNAL OF PARASITOLOGY 2015; 53:403-11. [PMID: 26323838 PMCID: PMC4566507 DOI: 10.3347/kjp.2015.53.4.403] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 07/23/2015] [Accepted: 07/23/2015] [Indexed: 11/23/2022]
Abstract
Plasmodium falciparum can invade all stages of red blood cells, while Plasmodium vivax can invade only reticulocytes. Although many P. vivax proteins have been discovered, their functions are largely unknown. Among them, P. vivax reticulocyte binding proteins (PvRBP1 and PvRBP2) recognize and bind to reticulocytes. Both proteins possess a C-terminal hydrophobic transmembrane domain, which drives adhesion to reticulocytes. PvRBP1 and PvRBP2 are large (> 326 kDa), which hinders identification of the functional domains. In this study, the complete genome information of the P. vivax RBP family was thoroughly analyzed using a prediction server with bioinformatics data to predict B-cell epitope domains. Eleven pvrbp family genes that included 2 pseudogenes and 9 full or partial length genes were selected and used to express recombinant proteins in a wheat germ cell-free system. The expressed proteins were used to evaluate the humoral immune response with vivax malaria patients and healthy individual serum samples by protein microarray. The recombinant fragments of 9 PvRBP proteins were successfully expressed; the soluble proteins ranged in molecular weight from 16 to 34 kDa. Evaluation of the humoral immune response to each recombinant PvRBP protein indicated a high antigenicity, with 38-88% sensitivity and 100% specificity. Of them, N-terminal parts of PvRBP2c (PVX_090325-1) and PvRBP2 like partial A (PVX_090330-1) elicited high antigenicity. In addition, the PvRBP2-like homologue B (PVX_116930) fragment was newly identified as high antigenicity and may be exploited as a potential antigenic candidate among the PvRBP family. The functional activity of the PvRBP family on merozoite invasion remains unknown.
Collapse
Affiliation(s)
- Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 200-701, Korea
| | - Jian Li
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 200-701, Korea.,Department of Parasitology, College of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Bo Wang
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 200-701, Korea.,Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Seong-Kyun Lee
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 200-701, Korea
| | - Myat Htut Nyunt
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 200-701, Korea.,Department of Medical Research, Yangon, Myanmar
| | - Sunghun Na
- Department of Obstetrics and Gynecology, School of Medicine, Kangwon National University, Chuncheon 200-701, Korea
| | - Jeong-Hyun Park
- Department of Anatomy, School of Medicine, Kangwon National University, Chuncheon 200-701, Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 200-701, Korea
| |
Collapse
|
42
|
Haase S, Zimmermann D, Olshina MA, Wilkinson M, Fisher F, Tan YH, Stewart RJ, Tonkin CJ, Wong W, Kovar DR, Baum J. Disassembly activity of actin-depolymerizing factor (ADF) is associated with distinct cellular processes in apicomplexan parasites. Mol Biol Cell 2015; 26:3001-12. [PMID: 26157165 PMCID: PMC4551315 DOI: 10.1091/mbc.e14-10-1427] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 06/30/2015] [Indexed: 12/15/2022] Open
Abstract
Complementation of a conditional KO of actin-depolymerizing factor (ADF) in Toxoplasma gondii demonstrates that ADF-dependent actin filament disassembly is essential for parasite development but not for cell motility. Furthermore, trans-genera complementation highlights genus-specific coevolution between ADF proteins and their native actins. Proteins of the actin-depolymerizing factor (ADF)/cofilin family have been shown to be crucial for the motility and survival of apicomplexan parasites. However, the mechanisms by which ADF proteins fulfill their function remain poorly understood. In this study, we investigate the comparative activities of ADF proteins from Toxoplasma gondii and Plasmodium falciparum, the human malaria parasite, using a conditional T. gondii ADF-knockout line complemented with ADF variants from either species. We show that P. falciparum ADF1 can fully restore native TgADF activity, demonstrating functional conservation between parasites. Strikingly, mutation of a key basic residue (Lys-72), previously implicated in disassembly in PfADF1, had no detectable phenotypic effect on parasite growth, motility, or development. In contrast, organelle segregation was severely impaired when complementing with a TgADF mutant lacking the corresponding residue (Lys-68). Biochemical analyses of each ADF protein confirmed the reduced ability of lysine mutants to mediate actin depolymerization via filament disassembly although not severing, in contrast to previous reports. These data suggest that actin filament disassembly is essential for apicomplexan parasite development but not for motility, as well as pointing to genus-specific coevolution between ADF proteins and their native actin.
Collapse
Affiliation(s)
- Silvia Haase
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Dennis Zimmermann
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| | - Maya A Olshina
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Mark Wilkinson
- Department of Life Sciences, Imperial College, London SW7 2AZ, United Kingdom
| | - Fabio Fisher
- Department of Life Sciences, Imperial College, London SW7 2AZ, United Kingdom
| | - Yan Hong Tan
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Rebecca J Stewart
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Christopher J Tonkin
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Wilson Wong
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - David R Kovar
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637
| | - Jake Baum
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia Department of Life Sciences, Imperial College, London SW7 2AZ, United Kingdom
| |
Collapse
|
43
|
Defining the morphology and mechanism of the hemoglobin transport pathway in Plasmodium falciparum-infected erythrocytes. EUKARYOTIC CELL 2015; 14:415-26. [PMID: 25724884 DOI: 10.1128/ec.00267-14] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 02/21/2015] [Indexed: 11/20/2022]
Abstract
Hemoglobin degradation during the asexual cycle of Plasmodium falciparum is an obligate process for parasite development and survival. It is established that hemoglobin is transported from the host erythrocyte to the parasite digestive vacuole (DV), but this biological process is not well characterized. Three-dimensional reconstructions made from serial thin-section electron micrographs of untreated, trophozoite-stage P. falciparum-infected erythrocytes (IRBC) or IRBC treated with different pharmacological agents provide new insight into the organization and regulation of the hemoglobin transport pathway. Hemoglobin internalization commences with the formation of cytostomes from localized, electron-dense collars at the interface of the parasite plasma and parasitophorous vacuolar membranes. The cytostomal collar does not function as a site of vesicle fission but rather serves to stabilize the maturing cytostome. We provide the first evidence that hemoglobin transport to the DV uses an actin-myosin motor system. Short-lived, hemoglobin-filled vesicles form from the distal end of the cytostomes through actin and dynamin-mediated processes. Results obtained with IRBC treated with N-ethylmaleimide (NEM) suggest that fusion of hemoglobin-containing vesicles with the DV may involve a soluble NEM-sensitive factor attachment protein receptor-dependent mechanism. In this report, we identify new key components of the hemoglobin transport pathway and provide a detailed characterization of its morphological organization and regulation.
Collapse
|
44
|
Kan A, Tan YH, Angrisano F, Hanssen E, Rogers KL, Whitehead L, Mollard VP, Cozijnsen A, Delves MJ, Crawford S, Sinden RE, McFadden GI, Leckie C, Bailey J, Baum J. Quantitative analysis of Plasmodium ookinete motion in three dimensions suggests a critical role for cell shape in the biomechanics of malaria parasite gliding motility. Cell Microbiol 2014; 16:734-50. [PMID: 24612056 PMCID: PMC4286792 DOI: 10.1111/cmi.12283] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 01/22/2014] [Accepted: 02/13/2014] [Indexed: 11/28/2022]
Abstract
Motility is a fundamental part of cellular life and survival, including for Plasmodium parasites--single-celled protozoan pathogens responsible for human malaria. The motile life cycle forms achieve motility, called gliding, via the activity of an internal actomyosin motor. Although gliding is based on the well-studied system of actin and myosin, its core biomechanics are not completely understood. Currently accepted models suggest it results from a specifically organized cellular motor that produces a rearward directional force. When linked to surface-bound adhesins, this force is passaged to the cell posterior, propelling the parasite forwards. Gliding motility is observed in all three life cycle stages of Plasmodium: sporozoites, merozoites and ookinetes. However, it is only the ookinetes--formed inside the midgut of infected mosquitoes--that display continuous gliding without the necessity of host cell entry. This makes them ideal candidates for invasion-free biomechanical analysis. Here we apply a plate-based imaging approach to study ookinete motion in three-dimensional (3D) space to understand Plasmodium cell motility and how movement facilitates midgut colonization. Using single-cell tracking and numerical analysis of parasite motion in 3D, our analysis demonstrates that ookinetes move with a conserved left-handed helical trajectory. Investigation of cell morphology suggests this trajectory may be based on the ookinete subpellicular cytoskeleton, with complementary whole and subcellular electron microscopy showing that, like their motion paths, ookinetes share a conserved left-handed corkscrew shape and underlying twisted microtubular architecture. Through comparisons of 3D movement between wild-type ookinetes and a cytoskeleton-knockout mutant we demonstrate that perturbation of cell shape changes motion from helical to broadly linear. Therefore, while the precise linkages between cellular architecture and actomyosin motor organization remain unknown, our analysis suggests that the molecular basis of cell shape may, in addition to motor force, be a key adaptive strategy for malaria parasite dissemination and, as such, transmission.
Collapse
Affiliation(s)
- Andrey Kan
- Victoria Research Laboratory, National ICT Australia (NICTA), Department of Computing and Information Systems, University of Melbourne, Melbourne, Vic., 3010, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Harding CR, Meissner M. The inner membrane complex through development of Toxoplasma gondii and Plasmodium. Cell Microbiol 2014; 16:632-41. [PMID: 24612102 PMCID: PMC4286798 DOI: 10.1111/cmi.12285] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 02/20/2014] [Accepted: 02/20/2014] [Indexed: 12/30/2022]
Abstract
Plasmodium spp. and Toxoplasma gondii are important human and veterinary pathogens. These parasites possess an unusual double membrane structure located directly below the plasma membrane named the inner membrane complex (IMC). First identified in early electron micrograph studies, huge advances in genetic manipulation of the Apicomplexa have allowed the visualization of a dynamic, highly structured cellular compartment with important roles in maintaining the structure and motility of these parasites. This review summarizes recent advances in the field and highlights the changes the IMC undergoes during the complex life cycles of the Apicomplexa.
Collapse
Affiliation(s)
- Clare R Harding
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, The University of Glasgow, Glasgow, UK
| | | |
Collapse
|
46
|
Affiliation(s)
- Gavin J. Wright
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
- Malaria Programme, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
- * E-mail: (GJW); (JCR)
| | - Julian C. Rayner
- Malaria Programme, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
- * E-mail: (GJW); (JCR)
| |
Collapse
|
47
|
Tate EW, Bell AS, Rackham MD, Wright MH. N-Myristoyltransferase as a potential drug target in malaria and leishmaniasis. Parasitology 2014; 141:37-49. [PMID: 23611109 DOI: 10.1017/s0031182013000450] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Infections caused by protozoan parasites are among the most widespread and intractable transmissible diseases affecting the developing world, with malaria and leishmaniasis being the most costly in terms of morbidity and mortality. Although new drugs are urgently required against both diseases in the face of ever-rising resistance to frontline therapies, very few candidates passing through development pipelines possess a known and novel mode of action. Set in the context of drugs currently in use and under development, we present the evidence for N-myristoyltransferase (NMT), an enzyme that N-terminally lipidates a wide range of specific target proteins through post-translational modification, as a potential drug target in malaria and the leishmaniases. We discuss the limitations of current knowledge regarding the downstream targets of this enzyme in protozoa, and our recent progress towards potent cell-active NMT inhibitors against the most clinically-relevant species of parasite. Finally, we outline the next steps required in terms of both tools to understand N-myristoylation in protozoan parasites, and the generation of potential development candidates based on the output of our recently-reported high-throughput screens.
Collapse
Affiliation(s)
- Edward W Tate
- Department of Chemistry, Institute of Chemical Biology, Imperial College London, London SW7 2AZ, UK
| | - Andrew S Bell
- Department of Chemistry, Institute of Chemical Biology, Imperial College London, London SW7 2AZ, UK
| | - Mark D Rackham
- Department of Chemistry, Institute of Chemical Biology, Imperial College London, London SW7 2AZ, UK
| | - Megan H Wright
- Department of Chemistry, Institute of Chemical Biology, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
48
|
Sharma P, Chitnis CE. Key molecular events during host cell invasion by Apicomplexan pathogens. Curr Opin Microbiol 2013; 16:432-7. [PMID: 23895827 DOI: 10.1016/j.mib.2013.07.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 07/03/2013] [Accepted: 07/04/2013] [Indexed: 10/26/2022]
Abstract
The ability of Apicomplexan parasites to invade host cells is key to their survival and pathogenesis. Plasmodium and Toxoplasma parasites share common mechanisms for invasion of host cells. Secretion of microneme and rhoptry proteins, tight junction formation and assembly of an acto-myosin motor are key steps for successful invasion by both parasites. Here, we review our understanding of the molecular basis for these steps.
Collapse
|
49
|
Ngwa CJ, Scheuermayer M, Mair GR, Kern S, Brügl T, Wirth CC, Aminake MN, Wiesner J, Fischer R, Vilcinskas A, Pradel G. Changes in the transcriptome of the malaria parasite Plasmodium falciparum during the initial phase of transmission from the human to the mosquito. BMC Genomics 2013; 14:256. [PMID: 23586929 PMCID: PMC3640944 DOI: 10.1186/1471-2164-14-256] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Accepted: 04/01/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The transmission of the malaria parasite Plasmodium falciparum from the human to the mosquito is mediated by dormant sexual precursor cells, the gametocytes, which become activated in the mosquito midgut. Because gametocytes are the only parasite stages able to establish an infection in the mosquito, they play a crucial role in spreading the tropical disease. The human-to-mosquito transmission triggers important molecular changes in the gametocytes, which initiate gametogenesis and prepare the parasite for life-cycle progression in the insect vector. RESULTS To better understand gene regulations during the initial phase of malaria parasite transmission, we focused on the transcriptome changes that occur within the first half hour of parasite development in the mosquito. Comparison of mRNA levels of P. falciparum gametocytes before and 30 min following activation using suppression subtractive hybridization (SSH) identified 126 genes, which changed in expression during gametogenesis. Among these, 17.5% had putative functions in signaling, 14.3% were assigned to cell cycle and gene expression, 8.7% were linked to the cytoskeleton or inner membrane complex, 7.9% were involved in proteostasis and 6.4% in metabolism, 12.7% were cell surface-associated proteins, 11.9% were assigned to other functions, and 20.6% represented genes of unknown function. For 40% of the identified genes there has as yet not been any protein evidence.For a subset of 27 genes, transcript changes during gametogenesis were studied in detail by real-time RT-PCR. Of these, 22 genes were expressed in gametocytes, and for 15 genes transcript expression in gametocytes was increased compared to asexual blood stage parasites. Transcript levels of seven genes were particularly high in activated gametocytes, pointing at functions downstream of gametocyte transmission to the mosquito. For selected genes, a regulated expression during gametogenesis was confirmed on the protein level, using quantitative confocal microscopy. CONCLUSIONS The obtained transcriptome data demonstrate the regulations of gene expression immediately following malaria parasite transmission to the mosquito. Our findings support the identification of proteins important for sexual reproduction and further development of the mosquito midgut stages and provide insights into the genetic basis of the rapid adaption of Plasmodium to the insect vector.
Collapse
Affiliation(s)
- Che Julius Ngwa
- Research Center for Infectious Diseases, University of Würzburg, Josef-Schneider-Strasse 2/D15, 97080 Würzburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Jones ML, Collins MO, Goulding D, Choudhary JS, Rayner JC. Analysis of protein palmitoylation reveals a pervasive role in Plasmodium development and pathogenesis. Cell Host Microbe 2013; 12:246-58. [PMID: 22901544 PMCID: PMC3501726 DOI: 10.1016/j.chom.2012.06.005] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 04/02/2012] [Accepted: 06/14/2012] [Indexed: 12/11/2022]
Abstract
Asexual stage Plasmodium falciparum replicates and undergoes a tightly regulated developmental process in human erythrocytes. One mechanism involved in the regulation of this process is posttranslational modification (PTM) of parasite proteins. Palmitoylation is a PTM in which cysteine residues undergo a reversible lipid modification, which can regulate target proteins in diverse ways. Using complementary palmitoyl protein purification approaches and quantitative mass spectrometry, we examined protein palmitoylation in asexual-stage P. falciparum parasites and identified over 400 palmitoylated proteins, including those involved in cytoadherence, drug resistance, signaling, development, and invasion. Consistent with the prevalence of palmitoylated proteins, palmitoylation is essential for P. falciparum asexual development and influences erythrocyte invasion by directly regulating the stability of components of the actin-myosin invasion motor. Furthermore, P. falciparum uses palmitoylation in diverse ways, stably modifying some proteins while dynamically palmitoylating others. Palmitoylation therefore plays a central role in regulating P. falciparum blood stage development.
Collapse
Affiliation(s)
- Matthew L Jones
- Malaria Programme, The Wellcome Trust Sanger Institute, The Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | | | | | | | | |
Collapse
|