1
|
Guizetti J. Imaging malaria parasites across scales and time. J Microsc 2025. [PMID: 39749880 DOI: 10.1111/jmi.13384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
The idea that disease is caused at the cellular level is so fundamental to us that we might forget the critical role microscopy played in generating and developing this insight. Visually identifying diseased or infected cells lays the foundation for any effort to curb human pathology. Since the discovery of the Plasmodium-infected red blood cells, which cause malaria, microscopy has undergone an impressive development now literally resolving individual molecules. This review explores the expansive field of light microscopy, focusing on its application to malaria research. Imaging technologies have transformed our understanding of biological systems, yet navigating the complex and ever-growing landscape of techniques can be daunting. This review offers a guide for researchers, especially those working on malaria, by providing historical context as well as practical advice on selecting the right imaging approach. The review advocates an integrated methodology that prioritises the research question while considering key factors like sample preparation, fluorophore choice, imaging modality, and data analysis. In addition to presenting seminal studies and innovative applications of microscopy, the review highlights a broad range of topics, from traditional techniques like white light microscopy to advanced methods such as superresolution microscopy and time-lapse imaging. It addresses the emerging challenges of microscopy, including phototoxicity and trade-offs in resolution and speed, and offers insights into future technologies that might impact malaria research. This review offers a mix of historical perspective, technological progress, and practical guidance that appeal to novice and advanced microscopists alike. It aims to inspire malaria researchers to explore imaging techniques that could enrich their studies, thus advancing the field through enhanced visual exploration of the parasite across scales and time.
Collapse
Affiliation(s)
- Julien Guizetti
- Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
2
|
Munjal A, Rex DAB, Garg P, Prasad TSK, Mishra SK, Malhotra Y, Yadav D, John J, P P, Rawal K, Singh S. Mass Spectrometric and Artificial Intelligence-Based Identification of the Secretome of Plasmodium falciparum Merozoites to Provide Novel Candidates for Vaccine Development Pipeline. Proteomics Clin Appl 2024; 18:e202300115. [PMID: 39082488 DOI: 10.1002/prca.202300115] [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: 11/02/2023] [Revised: 05/24/2024] [Accepted: 07/16/2024] [Indexed: 11/15/2024]
Abstract
PURPOSE Merozoites are the only extracellular form of blood stage parasites, making it a worthwhile target. Multiple invasins that are stored in the merozoite apical organelles, are secreted just prior to invasion, and mediates its interaction with RBC. A comprehensive identification of all these secreted invasins is lacking and this study addresses that gap. EXPERIMENTAL DESIGN Pf3D7 merozoites were enriched and triggered to discharge apical organelle contents by exposure to ionic conditions mimicking that of blood plasma. The secreted proteins were separated from cellular contents and both the fractions were subjected to proteomic analysis. Also, the identified secreted proteins were subjected to GO, PPI network analysis, and AI-based in silico approach to understand their vaccine candidacy. RESULTS A total of 63 proteins were identified in the secretory fraction with membrane and apical organellar localization. This includes various MSPs, micronemal EBAs and rhoptry bulb proteins, which play a crucial role in initial and late merozoite attachment, and majority of them qualified as vaccine candidates. CONCLUSION AND CLINICAL RELEVANCE We, for the first time, report the secretory repertoire of merozoite and its status for vaccine candidacy. This information can be utilized to develop better invasion blocking multisubunit vaccines, comprising of immunological epitopes from several secreted invasins.
Collapse
Affiliation(s)
- Akshay Munjal
- Special Centre of Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Devasahayam Arokia Balaya Rex
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
- Department of Laboratory Medicine and Pathology, Rochester, Minnesota, USA
| | - Prachi Garg
- Special Centre of Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | | | - Sai Kumar Mishra
- Center for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Noida, Uttar Pradesh, India
| | - Yuktika Malhotra
- Center for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Noida, Uttar Pradesh, India
| | - Deepika Yadav
- Center for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Noida, Uttar Pradesh, India
| | - Jerry John
- Center for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Noida, Uttar Pradesh, India
| | - Preeti P
- Center for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Noida, Uttar Pradesh, India
| | - Kamal Rawal
- Center for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Noida, Uttar Pradesh, India
| | - Shailja Singh
- Special Centre of Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
3
|
Yong JJM, Gao X, Prakash P, Ang JW, Lai SK, Chen MW, Neo JJL, Lescar J, Li HY, Preiser PR. Red blood cell signaling is functionally conserved in Plasmodium invasion. iScience 2024; 27:111052. [PMID: 39635131 PMCID: PMC11615254 DOI: 10.1016/j.isci.2024.111052] [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: 03/07/2023] [Revised: 06/20/2024] [Accepted: 09/24/2024] [Indexed: 12/07/2024] Open
Abstract
It is widely recognized that Plasmodium merozoites secrete ligands that interact with RBC receptors. Meanwhile the question on whether these interactions trigger RBC signals essential for invasion remains unresolved. There is evidence that Plasmodium falciparum parasites manipulate native RBC Ca2+ signaling to facilitate invasion. Here, we demonstrate a key role of RBC Ca2+ influx that is conserved across different Plasmodium species during invasion. RH5-basigin interaction triggers RBC cAMP increase to promote Ca2+ influx. The RBC signaling pathways can be blocked by a range of inhibitors during Plasmodium invasion, providing the evidence of a functionally conserved host cAMP-Ca2+ signaling that drives invasion and junction formation. Furthermore, RH5-basigin binding induces a pre-existing multimeric RBC membrane complex to undergo increased protein association containing the cAMP-inducing β-adrenergic receptor. Our work presents evidence of a conserved host cell signaling cascade necessary for Plasmodium invasion and will create opportunities to therapeutically target merozoite invasion.
Collapse
Affiliation(s)
- James Jia Ming Yong
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Xiaohong Gao
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Prem Prakash
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Jing Wen Ang
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Soak Kuan Lai
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Ming Wei Chen
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Jason Jun Long Neo
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Julien Lescar
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Hoi Yeung Li
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Peter R. Preiser
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| |
Collapse
|
4
|
Singh MK, Bonnell VA, Tojal Da Silva I, Santiago VF, Moraes MS, Adderley J, Doerig C, Palmisano G, Llinas M, Garcia CRS. A Plasmodium falciparum MORC protein complex modulates epigenetic control of gene expression through interaction with heterochromatin. eLife 2024; 12:RP92201. [PMID: 39412522 PMCID: PMC11483127 DOI: 10.7554/elife.92201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
Dynamic control of gene expression is critical for blood stage development of malaria parasites. Here, we used multi-omic analyses to investigate transcriptional regulation by the chromatin-associated microrchidia protein, MORC, during asexual blood stage development of the human malaria parasite Plasmodium falciparum. We show that PfMORC (PF3D7_1468100) interacts with a suite of nuclear proteins, including APETALA2 (ApiAP2) transcription factors (PfAP2-G5, PfAP2-O5, PfAP2-I, PF3D7_0420300, PF3D7_0613800, PF3D7_1107800, and PF3D7_1239200), a DNA helicase DS60 (PF3D7_1227100), and other chromatin remodelers (PfCHD1 and PfEELM2). Transcriptomic analysis of PfMORCHA-glmS knockdown parasites revealed 163 differentially expressed genes belonging to hypervariable multigene families, along with upregulation of genes mostly involved in host cell invasion. In vivo genome-wide chromatin occupancy analysis during both trophozoite and schizont stages of development demonstrates that PfMORC is recruited to repressed, multigene families, including the var genes in subtelomeric chromosomal regions. Collectively, we find that PfMORC is found in chromatin complexes that play a role in the epigenetic control of asexual blood stage transcriptional regulation and chromatin organization.
Collapse
Affiliation(s)
- Maneesh Kumar Singh
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São PauloSão PauloBrazil
| | - Victoria Ann Bonnell
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University ParkHarrisburgUnited States
- Huck Institutes Center for Eukaryotic Gene Regulation, Pennsylvania State University, University ParkHarrisburgUnited States
- Huck Institutes Center for Malaria Research, Pennsylvania State University, University ParkHarrisburgUnited States
| | | | | | - Miriam Santos Moraes
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São PauloSão PauloBrazil
| | - Jack Adderley
- School of Health and Biomedical Sciences, RMIT UniversityBundooraAustralia
| | - Christian Doerig
- School of Health and Biomedical Sciences, RMIT UniversityBundooraAustralia
| | - Giuseppe Palmisano
- Department of Parasitology, Institute of Biomedical Science, University of São PauloSão PauloBrazil
| | - Manuel Llinas
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University ParkHarrisburgUnited States
- Huck Institutes Center for Eukaryotic Gene Regulation, Pennsylvania State University, University ParkHarrisburgUnited States
- Huck Institutes Center for Malaria Research, Pennsylvania State University, University ParkHarrisburgUnited States
- Department of Chemistry, Pennsylvania State University, University ParkHarrisburgUnited States
| | - Celia RS Garcia
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São PauloSão PauloBrazil
| |
Collapse
|
5
|
Min H, Liang X, Wang C, Qin J, Boonhok R, Muneer A, Brashear AM, Li X, Minns AM, Adapa SR, Jiang RHY, Ning G, Cao Y, Lindner SE, Miao J, Cui L. The DEAD-box RNA helicase PfDOZI imposes opposing actions on RNA metabolism in Plasmodium falciparum. Nat Commun 2024; 15:3747. [PMID: 38702310 PMCID: PMC11068891 DOI: 10.1038/s41467-024-48140-4] [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: 02/04/2022] [Accepted: 04/19/2024] [Indexed: 05/06/2024] Open
Abstract
In malaria parasites, the regulation of mRNA translation, storage and degradation during development and life-stage transitions remains largely unknown. Here, we functionally characterized the DEAD-box RNA helicase PfDOZI in P. falciparum. Disruption of pfdozi enhanced asexual proliferation but reduced sexual commitment and impaired gametocyte development. By quantitative transcriptomics, we show that PfDOZI is involved in the regulation of invasion-related genes and sexual stage-specific genes during different developmental stages. PfDOZI predominantly participates in processing body-like mRNPs in schizonts but germ cell granule-like mRNPs in gametocytes to impose opposing actions of degradation and protection on different mRNA targets. We further show the formation of stress granule-like mRNPs during nutritional deprivation, highlighting an essential role of PfDOZI-associated mRNPs in stress response. We demonstrate that PfDOZI participates in distinct mRNPs to maintain mRNA homeostasis in response to life-stage transition and environmental changes by differentially executing post-transcriptional regulation on the target mRNAs.
Collapse
Affiliation(s)
- Hui Min
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL, 33612, USA
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Xiaoying Liang
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL, 33612, USA
| | - Chengqi Wang
- Center for Global Health and Infectious Diseases, Department of Global Health, College of Public Health, University of South Florida, Tampa, FL, 33612, USA
| | - Junling Qin
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL, 33612, USA
| | - Rachasak Boonhok
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL, 33612, USA
- Department of Medical Technology, School of Allied Health Sciences, and Research Excellence Center for Innovation and Health Products (RECIHP), Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | - Azhar Muneer
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL, 33612, USA
| | - Awtum M Brashear
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL, 33612, USA
| | - Xiaolian Li
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL, 33612, USA
| | - Allen M Minns
- Department of Biochemistry and Molecular Biology, Huck Center for Malaria Research, Pennsylvania State University, University Park, PA, 16802, USA
| | - Swamy Rakesh Adapa
- Center for Global Health and Infectious Diseases, Department of Global Health, College of Public Health, University of South Florida, Tampa, FL, 33612, USA
| | - Rays H Y Jiang
- Center for Global Health and Infectious Diseases, Department of Global Health, College of Public Health, University of South Florida, Tampa, FL, 33612, USA
| | - Gang Ning
- Electron Microscopy Facility, The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Scott E Lindner
- Department of Biochemistry and Molecular Biology, Huck Center for Malaria Research, Pennsylvania State University, University Park, PA, 16802, USA
| | - Jun Miao
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL, 33612, USA.
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL, 33612, USA.
| |
Collapse
|
6
|
Desai SA. Novel Ion Channel Genes in Malaria Parasites. Genes (Basel) 2024; 15:296. [PMID: 38540355 PMCID: PMC10970509 DOI: 10.3390/genes15030296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 06/14/2024] Open
Abstract
Ion channels serve many cellular functions including ion homeostasis, volume regulation, signaling, nutrient acquisition, and developmental progression. Although the complex life cycles of malaria parasites necessitate ion and solute flux across membranes, the whole-genome sequencing of the human pathogen Plasmodium falciparum revealed remarkably few orthologs of known ion channel genes. Contrasting with this, biochemical studies have implicated the channel-mediated flux of ions and nutritive solutes across several membranes in infected erythrocytes. Here, I review advances in the cellular and molecular biology of ion channels in malaria parasites. These studies have implicated novel parasite genes in the formation of at least two ion channels, with additional ion channels likely present in various membranes and parasite stages. Computational approaches that rely on homology to known channel genes from higher organisms will not be very helpful in identifying the molecular determinants of these activities. Given their unusual properties, novel molecular and structural features, and essential roles in pathogen survival and development, parasite channels should be promising targets for therapy development.
Collapse
Affiliation(s)
- Sanjay A Desai
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| |
Collapse
|
7
|
Sitaraman R. Subversion from Within and Without: Effector Molecule Transfer from Obligate Intracellular Apicomplexan Parasites to Human Host Cells. Results Probl Cell Differ 2024; 73:521-535. [PMID: 39242391 DOI: 10.1007/978-3-031-62036-2_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2024]
Abstract
Intracellular protozoan pathogens have to negotiate the internal environment of the host cell they find themselves in, as well as manipulate the host cell to ensure their own survival, replication, and dissemination. The transfer of key effector molecules from the pathogen to the host cell is crucial to this interaction and is technically more demanding to study as compared to an extracellular pathogen. While several effector molecules have been identified, the mechanisms and conditions underlying their transfer to the host cell remain partly or entirely unknown. Improvements in experimental systems have revealed tantalizing details of such intercellular transfer, which form the subject of this chapter.
Collapse
|
8
|
Saeed S, Tremp AZ, Dessens JT. Plasmodium sporozoite excystation involves local breakdown of the oocyst capsule. Sci Rep 2023; 13:22222. [PMID: 38097730 PMCID: PMC10721906 DOI: 10.1038/s41598-023-49442-1] [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: 08/03/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023] Open
Abstract
Plasmodium oocysts develop on the abluminal side of the mosquito midgut in relatively small numbers. Oocysts possess an extracellular cell wall-the capsule-to protect them from the insect's haemolymph environment. To further maximise transmission, each oocyst generates hundreds of sporozoites through an asexual multiplication step called sporogony. Completion of transmission requires sporozoite egress from the capsule (excystation), but this process remains poorly understood. In this study, we fused the parasite-encoded capsule protein Cap380 with green fluorescent protein in a transgenic P. berghei line, allowing live fluorescence imaging of capsules throughout sporogony and sporozoite excystation. The results show that capsules progressively weaken during sporulation ultimately resulting in sporozoite exit through small holes. Prior to formation of the holes, local thinning of the capsule was observed. Our findings support an excystation model based on local, rather than global, weakening of the capsule likely facilitated by local re-orientation of sporozoites and apical secretion.
Collapse
Affiliation(s)
- Sadia Saeed
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Annie Z Tremp
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Johannes T Dessens
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London, WC1E 7HT, UK.
| |
Collapse
|
9
|
Valleau D, Sidik SM, Godoy LC, Acevedo‐Sánchez Y, Pasaje CFA, Huynh M, Carruthers VB, Niles JC, Lourido S. A conserved complex of microneme proteins mediates rhoptry discharge in Toxoplasma. EMBO J 2023; 42:e113155. [PMID: 37886905 PMCID: PMC10690463 DOI: 10.15252/embj.2022113155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/28/2023] Open
Abstract
Apicomplexan parasites discharge specialized organelles called rhoptries upon host cell contact to mediate invasion. The events that drive rhoptry discharge are poorly understood, yet essential to sustain the apicomplexan parasitic life cycle. Rhoptry discharge appears to depend on proteins secreted from another set of organelles called micronemes, which vary in function from allowing host cell binding to facilitation of gliding motility. Here we examine the function of the microneme protein CLAMP, which we previously found to be necessary for Toxoplasma gondii host cell invasion, and demonstrate its essential role in rhoptry discharge. CLAMP forms a distinct complex with two other microneme proteins, the invasion-associated SPATR, and a previously uncharacterized protein we name CLAMP-linked invasion protein (CLIP). CLAMP deficiency does not impact parasite adhesion or microneme protein secretion; however, knockdown of any member of the CLAMP complex affects rhoptry discharge. Phylogenetic analysis suggests orthologs of the essential complex components, CLAMP and CLIP, are ubiquitous across apicomplexans. SPATR appears to act as an accessory factor in Toxoplasma, but despite incomplete conservation is also essential for invasion during Plasmodium falciparum blood stages. Together, our results reveal a new protein complex that mediates rhoptry discharge following host-cell contact.
Collapse
Affiliation(s)
| | | | - Luiz C Godoy
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
| | | | | | - My‐Hang Huynh
- Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Vern B Carruthers
- Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Jacquin C Niles
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Sebastian Lourido
- Whitehead InstituteCambridgeMAUSA
- Biology DepartmentMassachusetts Institute of TechnologyCambridgeMAUSA
| |
Collapse
|
10
|
Andrews M, Baum J, Gilson PR, Wilson DW. Bottoms up! Malaria parasite invasion the right way around. Trends Parasitol 2023; 39:1004-1013. [PMID: 37827961 DOI: 10.1016/j.pt.2023.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 10/14/2023]
Abstract
A critical part of the malaria parasite's life cycle is invasion of red blood cells (RBCs) by merozoites. Inside RBCs, the parasite forms a schizont, which undergoes segmentation to produce daughter merozoites. These cells are released, establishing cycles of invasion. Traditionally, merozoites are represented as nonmotile, egg-shaped cells that invade RBCs 'narrower end' first and pack within schizonts with this narrower end facing outwards. Here, we discuss recent evidence and re-evaluate previous data which suggest that merozoites are capable of motility and have spherical or elongated-teardrop shapes. Furthermore, merozoites invade RBCs 'wider end' first and pack within schizonts with this wider end facing outwards. We encourage the field to review this revised model and consider its implications for future studies.
Collapse
Affiliation(s)
- Mia Andrews
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide 5005, Australia
| | - Jake Baum
- School of Biomedical Sciences, UNSW Sydney, Kensington, NSW 2052, Australia; Department of Life Sciences, Imperial College London, South Kensington, London SW7 2AZ, UK
| | - Paul R Gilson
- Burnet Institute, Melbourne 3004, Victoria, Australia; Department of Microbiology and Immunology, The University of Melbourne, Melbourne 3010, Victoria, Australia
| | - Danny W Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide 5005, Australia; Burnet Institute, Melbourne 3004, Victoria, Australia; Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, 5005, SA, Australia.
| |
Collapse
|
11
|
Shunmugam S, Quansah N, Flammersfeld A, Islam MM, Sassmannshausen J, Bennink S, Yamaryo-Botté Y, Pradel G, Botté CY. The patatin-like phospholipase PfPNPLA2 is involved in the mitochondrial degradation of phosphatidylglycerol during Plasmodium falciparum blood stage development. Front Cell Infect Microbiol 2023; 13:997245. [PMID: 38089812 PMCID: PMC10711835 DOI: 10.3389/fcimb.2023.997245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/09/2023] [Indexed: 12/18/2023] Open
Abstract
Plasmodium falciparum is an Apicomplexa responsible for human malaria, a major disease causing more than ½ million deaths every year, against which there is no fully efficient vaccine. The current rapid emergence of drug resistances emphasizes the need to identify novel drug targets. Increasing evidences show that lipid synthesis and trafficking are essential for parasite survival and pathogenesis, and that these pathways represent potential points of attack. Large amounts of phospholipids are needed for the generation of membrane compartments for newly divided parasites in the host cell. Parasite membrane homeostasis is achieved by an essential combination of parasite de novo lipid synthesis/recycling and massive host lipid scavenging. Latest data suggest that the mobilization and channeling of lipid resources is key for asexual parasite survival within the host red blood cell, but the molecular actors allowing lipid acquisition are poorly characterized. Enzymes remodeling lipids such as phospholipases are likely involved in these mechanisms. P. falciparum possesses an unusually large set of phospholipases, whose functions are largely unknown. Here we focused on the putative patatin-like phospholipase PfPNPLA2, for which we generated an glmS-inducible knockdown line and investigated its role during blood stages malaria. Disruption of the mitochondrial PfPNPLA2 in the asexual blood stages affected mitochondrial morphology and further induced a significant defect in parasite replication and survival, in particular under low host lipid availability. Lipidomic analyses revealed that PfPNPLA2 specifically degrades the parasite membrane lipid phosphatidylglycerol to generate lysobisphosphatidic acid. PfPNPLA2 knockdown further resulted in an increased host lipid scavenging accumulating in the form of storage lipids and free fatty acids. These results suggest that PfPNPLA2 is involved in the recycling of parasite phosphatidylglycerol to sustain optimal intraerythrocytic development when the host resources are scarce. This work strengthens our understanding of the complex lipid homeostasis pathways to acquire lipids and allow asexual parasite survival.
Collapse
Affiliation(s)
- Serena Shunmugam
- Apicolipid Team, Institute for Avanced Biosciences, Centre National pour la Recherche Scientifique (CNRS) UMR5309, Institut National de la Santé et de la Recherche Médicale, Université Grenoble Alpes, Grenoble, France
| | - Nyamekye Quansah
- Apicolipid Team, Institute for Avanced Biosciences, Centre National pour la Recherche Scientifique (CNRS) UMR5309, Institut National de la Santé et de la Recherche Médicale, Université Grenoble Alpes, Grenoble, France
| | - Ansgar Flammersfeld
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Md Muzahidul Islam
- International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Juliane Sassmannshausen
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Sandra Bennink
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Yoshiki Yamaryo-Botté
- Apicolipid Team, Institute for Avanced Biosciences, Centre National pour la Recherche Scientifique (CNRS) UMR5309, Institut National de la Santé et de la Recherche Médicale, Université Grenoble Alpes, Grenoble, France
| | - Gabriele Pradel
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Cyrille Y. Botté
- Apicolipid Team, Institute for Avanced Biosciences, Centre National pour la Recherche Scientifique (CNRS) UMR5309, Institut National de la Santé et de la Recherche Médicale, Université Grenoble Alpes, Grenoble, France
| |
Collapse
|
12
|
Gupta S, Saini M, Joshi N, Shafi S, Najmi AK, Singh S. Antimalarial and Plasmodium falciparum serpentine receptor 12 targeting effect of FDA approved purinergic receptor antagonist. J Biomol Struct Dyn 2023; 41:9462-9475. [PMID: 36351236 DOI: 10.1080/07391102.2022.2142298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022]
Abstract
Intraerythrocytic stages of Plasmodium falciparum responsible for all clinical manifestations of malaria are regulated by array of signalling cascades that represent attractive targets for antimalarial therapy. G-protein coupled receptors (GPCRs) are druggable targets in the treatment of various pathological conditions, however, there is limited understanding about the role of GPCRs in malaria pathogenesis. In Plasmodium, serpentine receptors (PfSR1, PfSR10, PfSR12 and PfSR25) with GPCR-like membrane topology have been reported with the finite knowledge about their potential as antimalarial targets. We analyzed the localization of these receptors in malaria parasite by immunofluorescence assays. All four receptors were expressed in blood stages with PfSR12 expressing more in late intraerythrocytic stages. Further, we evaluated the druggability of PfSR12 using FDA-approved P2Y purinergic receptor antagonist, Prasugrel and its active metabolite R138727, which is proposed to be specific towards PfSR12. Interestingly, biophysical analysis indicated strong binding between PfSR12 and R138727 as compared to the prodrug Prasugrel. This binding interaction was further confirmed by thermal shift assay. Treatment of parasite with Prasugrel and R138727 resulted in growth inhibition of P. falciparum indicating an important role of purinergic signalling and PfSR12 in parasite survival. Next, progression studies indicated the inhibitory effect of Prasugrel begins in late erythrocyte stages corroborating with PfSR12 expression at these stages. Furthermore, Prasugrel also blocked in vivo growth of malaria parasite in a mouse experimental model. This study indicates the presence of P2Y type of purinergic signalling in growth and development of malaria parasite and suggests PfSR12, putative purinergic receptor druggability through Prasugrel.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Sonal Gupta
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Monika Saini
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
- Department of Life Sciences, Shiv Nadar University, Gautam Buddha Nagar, India
| | - Nishant Joshi
- Department of Life Sciences, Shiv Nadar University, Gautam Buddha Nagar, India
| | - Sadat Shafi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard University, New Delhi, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard University, New Delhi, India
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
13
|
Shortt E, Hackett CG, Stadler RV, Kent RS, Herneisen AL, Ward GE, Lourido S. CDPK2A and CDPK1 form a signaling module upstream of Toxoplasma motility. mBio 2023; 14:e0135823. [PMID: 37610220 PMCID: PMC10653799 DOI: 10.1128/mbio.01358-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 06/17/2023] [Indexed: 08/24/2023] Open
Abstract
IMPORTANCE This work uncovers interactions between various signaling pathways that govern Toxoplasma gondii egress. Specifically, we compare the function of three canonical calcium-dependent protein kinases (CDPKs) using chemical-genetic and conditional-depletion approaches. We describe the function of a previously uncharacterized CDPK, CDPK2A, in the Toxoplasma lytic cycle, demonstrating that it contributes to parasite fitness through regulation of microneme discharge, gliding motility, and egress from infected host cells. Comparison of analog-sensitive kinase alleles and conditionally depleted alleles uncovered epistasis between CDPK2A and CDPK1, implying a partial functional redundancy. Understanding the topology of signaling pathways underlying key events in the parasite life cycle can aid in efforts targeting kinases for anti-parasitic therapies.
Collapse
Affiliation(s)
- Emily Shortt
- Whitehead Institute, Cambridge, Massachusetts, USA
| | | | - Rachel V. Stadler
- Department of Microbiology and Molecular Genetics, University of Vermont Larner College of Medicine, Burlington, Vermont, USA
| | - Robyn S. Kent
- Department of Microbiology and Molecular Genetics, University of Vermont Larner College of Medicine, Burlington, Vermont, USA
| | - Alice L. Herneisen
- Whitehead Institute, Cambridge, Massachusetts, USA
- Biology Department, MIT, Cambridge, Massachusetts, USA
| | - Gary E. Ward
- Department of Microbiology and Molecular Genetics, University of Vermont Larner College of Medicine, Burlington, Vermont, USA
| | - Sebastian Lourido
- Whitehead Institute, Cambridge, Massachusetts, USA
- Biology Department, MIT, Cambridge, Massachusetts, USA
| |
Collapse
|
14
|
Arabiotorre A, Bankaitis VA, Grabon A. Regulation of phosphoinositide metabolism in Apicomplexan parasites. Front Cell Dev Biol 2023; 11:1163574. [PMID: 37791074 PMCID: PMC10543664 DOI: 10.3389/fcell.2023.1163574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 07/11/2023] [Indexed: 10/05/2023] Open
Abstract
Phosphoinositides are a biologically essential class of phospholipids that contribute to organelle membrane identity, modulate membrane trafficking pathways, and are central components of major signal transduction pathways that operate on the cytosolic face of intracellular membranes in eukaryotes. Apicomplexans (such as Toxoplasma gondii and Plasmodium spp.) are obligate intracellular parasites that are important causative agents of disease in animals and humans. Recent advances in molecular and cell biology of Apicomplexan parasites reveal important roles for phosphoinositide signaling in key aspects of parasitosis. These include invasion of host cells, intracellular survival and replication, egress from host cells, and extracellular motility. As Apicomplexans have adapted to the organization of essential signaling pathways to accommodate their complex parasitic lifestyle, these organisms offer experimentally tractable systems for studying the evolution, conservation, and repurposing of phosphoinositide signaling. In this review, we describe the regulatory mechanisms that control the spatial and temporal regulation of phosphoinositides in the Apicomplexan parasites Plasmodium and T. gondii. We further discuss the similarities and differences presented by Apicomplexan phosphoinositide signaling relative to how these pathways are regulated in other eukaryotic organisms.
Collapse
Affiliation(s)
- Angela Arabiotorre
- Department of Cell Biology and Genetics, College of Medicine Texas A&M Health Sciences Center College Station, Bryan, TX, United States
| | - Vytas A. Bankaitis
- Department of Cell Biology and Genetics, College of Medicine Texas A&M Health Sciences Center College Station, Bryan, TX, United States
- Department of Biochemistry and Biophysics Texas A&M University College Station, College Station, TX, United States
- Department of Chemistry Texas A&M University College Station, College Station, TX, United States
| | - Aby Grabon
- Department of Cell Biology and Genetics, College of Medicine Texas A&M Health Sciences Center College Station, Bryan, TX, United States
| |
Collapse
|
15
|
Burda PC, Ramaprasad A, Bielfeld S, Pietsch E, Woitalla A, Söhnchen C, Singh MN, Strauss J, Sait A, Collinson LM, Schwudke D, Blackman MJ, Gilberger TW. Global analysis of putative phospholipases in Plasmodium falciparum reveals an essential role of the phosphoinositide-specific phospholipase C in parasite maturation. mBio 2023; 14:e0141323. [PMID: 37489900 PMCID: PMC10470789 DOI: 10.1128/mbio.01413-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 07/26/2023] Open
Abstract
For its replication within red blood cells, the malaria parasite depends on a highly active and regulated lipid metabolism. Enzymes involved in lipid metabolic processes such as phospholipases are, therefore, potential drug targets. Here, using reverse genetics approaches, we show that only 1 out of the 19 putative phospholipases expressed in asexual blood stages of Plasmodium falciparum is essential for proliferation in vitro, pointing toward a high level of redundancy among members of this enzyme family. Using conditional mislocalization and gene disruption techniques, we show that this essential phosphoinositide-specific phospholipase C (PI-PLC, PF3D7_1013500) has a previously unrecognized essential role during intracellular parasite maturation, long before its previously perceived role in parasite egress and invasion. Subsequent lipidomic analysis suggests that PI-PLC mediates cleavage of phosphatidylinositol bisphosphate (PIP2) in schizont-stage parasites, underlining its critical role in regulating phosphoinositide levels in the parasite. IMPORTANCE The clinical symptoms of malaria arise due to repeated rounds of replication of Plasmodium parasites within red blood cells (RBCs). Central to this is an intense period of membrane biogenesis. Generation of membranes not only requires de novo synthesis and acquisition but also the degradation of phospholipids, a function that is performed by phospholipases. In this study, we investigate the essentiality of the 19 putative phospholipase enzymes that the human malaria parasite Plasmodium falciparum expresses during its replication within RBCs. We not only show that a high level of functional redundancy exists among these enzymes but, at the same time, also identify an essential role for the phosphoinositide-specific phospholipase C in parasite development and cleavage of the phospholipid phosphatidylinositol bisphosphate.
Collapse
Affiliation(s)
- Paul-Christian Burda
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- University of Hamburg, Hamburg, Germany
| | - Abhinay Ramaprasad
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Sabrina Bielfeld
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- University of Hamburg, Hamburg, Germany
| | - Emma Pietsch
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- University of Hamburg, Hamburg, Germany
| | - Anna Woitalla
- Division of Bioanalytical Chemistry, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Christoph Söhnchen
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- University of Hamburg, Hamburg, Germany
| | - Mehar Nihal Singh
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Jan Strauss
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- University of Hamburg, Hamburg, Germany
| | - Aaron Sait
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Lucy M. Collinson
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Dominik Schwudke
- Division of Bioanalytical Chemistry, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
- German Center for Infection Research, Thematic Translational Unit Tuberculosis, Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel, Germany
- German Center for Lung Research (DZL), Airway Research Center North (ARCN), Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Michael J. Blackman
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Tim-Wolf Gilberger
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- University of Hamburg, Hamburg, Germany
| |
Collapse
|
16
|
Hart MN, Mohring F, DonVito SM, Thomas JA, Muller-Sienerth N, Wright GJ, Knuepfer E, Saibil HR, Moon RW. Sequential roles for red blood cell binding proteins enable phased commitment to invasion for malaria parasites. Nat Commun 2023; 14:4619. [PMID: 37528099 PMCID: PMC10393984 DOI: 10.1038/s41467-023-40357-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 07/20/2023] [Indexed: 08/03/2023] Open
Abstract
Invasion of red blood cells (RBCs) by Plasmodium merozoites is critical to their continued survival within the host. Two major protein families, the Duffy binding-like proteins (DBPs/EBAs) and the reticulocyte binding like proteins (RBLs/RHs) have been studied extensively in P. falciparum and are hypothesized to have overlapping, but critical roles just prior to host cell entry. The zoonotic malaria parasite, P. knowlesi, has larger invasive merozoites and contains a smaller, less redundant, DBP and RBL repertoire than P. falciparum. One DBP (DBPα) and one RBL, normocyte binding protein Xa (NBPXa) are essential for invasion of human RBCs. Taking advantage of the unique biological features of P. knowlesi and iterative CRISPR-Cas9 genome editing, we determine the precise order of key invasion milestones and demonstrate distinct roles for each family. These distinct roles support a mechanism for phased commitment to invasion and can be targeted synergistically with invasion inhibitory antibodies.
Collapse
Affiliation(s)
- Melissa N Hart
- Department of Infection Biology, Faculty of Infectious and Tropical Disease, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
- Department of Pathobiology and Population Sciences, Royal Veterinary College, Hawkshead Lane, Hatfield, AL9 7TA, UK
| | - Franziska Mohring
- Department of Infection Biology, Faculty of Infectious and Tropical Disease, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Sophia M DonVito
- Department of Infection Biology, Faculty of Infectious and Tropical Disease, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - James A Thomas
- Department of Infection Biology, Faculty of Infectious and Tropical Disease, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | | | - Gavin J Wright
- Wellcome Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York, Wentworth Way, York, YO10 5DD, UK
| | - Ellen Knuepfer
- Department of Pathobiology and Population Sciences, Royal Veterinary College, Hawkshead Lane, Hatfield, AL9 7TA, UK
- Malaria Parasitology Laboratory, Francis Crick Institute, London, NW1 1AT, UK
| | - Helen R Saibil
- ISMB, Biological Sciences, Birkbeck, University of London, Malet St, London, WC1E 7HX, UK
| | - Robert W Moon
- Department of Infection Biology, Faculty of Infectious and Tropical Disease, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK.
| |
Collapse
|
17
|
Vallintine T, van Ooij C. Timing of dense granule biogenesis in asexual malaria parasites. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001389. [PMID: 37647112 PMCID: PMC10482371 DOI: 10.1099/mic.0.001389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/15/2023] [Indexed: 09/01/2023]
Abstract
Malaria is an important infectious disease that continues to claim hundreds of thousands of lives annually. The disease is caused by infection of host erythrocytes by apicomplexan parasites of the genus Plasmodium. The parasite contains three different apical organelles - micronemes, rhoptries and dense granules (DGs) - whose contents are secreted to mediate binding to and invasion of the host cell and the extensive remodelling of the host cell that occurs following invasion. Whereas the roles of micronemes and rhoptries in binding and invasion of the host erythrocyte have been studied in detail, the roles of DGs in Plasmodium parasites are poorly understood. They have been proposed to control host cell remodelling through regulated protein secretion after invasion, but many basic aspects of the biology of DGs remain unknown. Here we describe DG biogenesis timing for the first time, using RESA localization as a proxy for the timing of DG formation. We show that DG formation commences approximately 37 min prior to schizont egress, as measured by the recruitment of the DG marker RESA. Furthermore, using a bioinformatics approach, we aimed to predict additional cargo of the DGs and identified the J-dot protein HSP40 as a DG protein, further supporting the very early role of these organelles in the interaction of the parasite with the host cell.
Collapse
Affiliation(s)
- Tansy Vallintine
- Faculty of Infectious Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Christiaan van Ooij
- Faculty of Infectious Diseases, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
18
|
Santos BMD, Przyborski JM, Garcia CRS. Changes in K + Concentration as a Signaling Mechanism in the Apicomplexa Parasites Plasmodium and Toxoplasma. Int J Mol Sci 2023; 24:ijms24087276. [PMID: 37108438 PMCID: PMC10138558 DOI: 10.3390/ijms24087276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/04/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
During their life cycle, apicomplexan parasites pass through different microenvironments and encounter a range of ion concentrations. The discovery that the GPCR-like SR25 in Plasmodium falciparum is activated by a shift in potassium concentration indicates that the parasite can take advantage of its development by sensing different ionic concentrations in the external milieu. This pathway involves the activation of phospholipase C and an increase in cytosolic calcium. In the present report, we summarize the information available in the literature regarding the role of potassium ions during parasite development. A deeper understanding of the mechanisms that allow the parasite to cope with ionic potassium changes contributes to our knowledge about the cell cycle of Plasmodium spp.
Collapse
Affiliation(s)
- Benedito M Dos Santos
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Jude M Przyborski
- Department of Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus-Liebig University, 35390 Gießen, Germany
| | - Célia R S Garcia
- Department of Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus-Liebig University, 35390 Gießen, Germany
| |
Collapse
|
19
|
Msosa C, Abdalrahman T, Franz T. An analytical model describing the mechanics of erythrocyte membrane wrapping during active invasion of a plasmodium falciparum merozoite. J Mech Behav Biomed Mater 2023; 140:105685. [PMID: 36746046 DOI: 10.1016/j.jmbbm.2023.105685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 12/22/2022] [Accepted: 01/23/2023] [Indexed: 01/27/2023]
Abstract
The invasion of a merozoite into an erythrocyte by membrane wrapping is a hallmark of malaria pathogenesis. The invasion involves biomechanical interactions whereby the merozoite exerts actomyosin-based forces to push itself into and through the erythrocyte membrane while concurrently inducing biochemical damage to the erythrocyte membrane. Whereas the biochemical damage process has been investigated, the detailed mechanistic understanding of the invasion mechanics remains limited. Thus, the current study aimed to develop a mathematical model describing the mechanical factors involved in the merozoite invasion into an erythrocyte and explore the invasion mechanics. A shell theory model was developed comprising constitutive, equilibrium and governing equations of the deformable erythrocyte membrane to predict membrane mechanics during the wrapping of an entire non-deformable ellipsoidal merozoite. Predicted parameters include principal erythrocyte membrane deformations and stresses, wrapping and indentation forces, and indentation work. The numerical investigations considered two limits for the erythrocyte membrane deformation during wrapping (4% and 51% areal strain) and erythrocyte membrane phosphorylation (decrease of membrane elastic modulus from 1 to 0.5 kPa). For an intact erythrocyte, the maximum indentation force was 1 and 8.5 pN, and the indentation work was 1.92 × 10-18 and 1.40 × 10-17 J for 4% and 51% areal membrane strain. Phosphorylation damage in the erythrocyte membrane reduced the required indentation work by 50% to 0.97 × 10-18 and 0.70 × 10-17 J for 4% and 51% areal strain. The current study demonstrated the developed model's feasibility to provide new knowledge on the physical mechanisms of the merozoite invasion process that contribute to the invasion efficiency towards the discovery of new invasion-blocking anti-malaria drugs.
Collapse
Affiliation(s)
- Chimwemwe Msosa
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Observatory, 7925, South Africa; Faculty of Engineering, Department of Electrical Engineering, Malawi University of Business and Applied Sciences, Blantyre, Malawi.
| | - Tamer Abdalrahman
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Observatory, 7925, South Africa; Computational Mechanobiology, Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité Universitätsmedizin, Berlin, 13353, Germany
| | - Thomas Franz
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Observatory, 7925, South Africa; Bioengineering Science Research Group, Engineering Sciences, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO171BJ, UK
| |
Collapse
|
20
|
Elsworth B, Keroack C, Rezvani Y, Paul A, Barazorda K, Tennessen J, Sack S, Moreira C, Gubbels MJ, Meyers M, Zarringhalam K, Duraisingh M. Babesia divergens egress from host cells is orchestrated by essential and druggable kinases and proteases. RESEARCH SQUARE 2023:rs.3.rs-2553721. [PMID: 36909484 PMCID: PMC10002801 DOI: 10.21203/rs.3.rs-2553721/v1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Apicomplexan egress from host cells is fundamental to the spread of infection and is poorly characterized in Babesia spp., parasites of veterinary importance and emerging zoonoses. Through the use of video microscopy, transcriptomics and chemical genetics, we have implicated signaling, proteases and gliding motility as key drivers of egress by Babesia divergens. We developed reverse genetics to perform a knockdown screen of putative mediators of egress, identifying kinases and proteases involved in distinct steps of egress (ASP3, PKG and CDPK4) and invasion (ASP2, ASP3 and PKG). Inhibition of egress leads to continued intracellular replication, indicating exit from the replication cycle is uncoupled from egress. Chemical genetics validated PKG, ASP2 and ASP3 as druggable targets in Babesia spp. All taken together, egress in B. divergens more closely resembles T. gondii than the more evolutionarily-related Plasmodium spp. We have established a molecular framework for biological and translational studies of B. divergens egress.
Collapse
|
21
|
Defining species-specific and conserved interactions of apical membrane protein 1 during erythrocyte invasion in malaria to inform multi-species vaccines. Cell Mol Life Sci 2023; 80:74. [PMID: 36847896 PMCID: PMC9969379 DOI: 10.1007/s00018-023-04712-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/17/2023] [Accepted: 01/30/2023] [Indexed: 03/01/2023]
Abstract
Plasmodium falciparum and P. vivax are the major causes of human malaria, and P. knowlesi is an important additional cause in SE Asia. Binding of apical membrane antigen 1 (AMA1) to rhoptry neck protein 2 (RON2) was thought to be essential for merozoite invasion of erythrocytes by Plasmodium spp. Our findings reveal that P. falciparum and P. vivax have diverged and show species-specific binding of AMA1 to RON2, determined by a β-hairpin loop in RON2 and specific residues in AMA1 Loop1E. In contrast, cross-species binding of AMA1 to RON2 is retained between P. vivax and P. knowlesi. Mutation of specific amino acids in AMA1 Loop1E in P. falciparum or P. vivax ablated RON2 binding without impacting erythrocyte invasion. This indicates that the AMA1-RON2-loop interaction is not essential for invasion and additional AMA1 interactions are involved. Mutations in AMA1 that disrupt RON2 binding also enable escape of invasion inhibitory antibodies. Therefore, vaccines and therapeutics will need to be broader than targeting only the AMA1-RON2 interaction. Antibodies targeting AMA1 domain 3 had greater invasion-inhibitory activity when RON2-loop binding was ablated, suggesting this domain is a promising additional target for vaccine development. Targeting multiple AMA1 interactions involved in invasion may enable vaccines that generate more potent inhibitory antibodies and address the capacity for immune evasion. Findings on specific residues for invasion function and species divergence and conservation can inform novel vaccines and therapeutics against malaria caused by three species, including the potential for cross-species vaccines.
Collapse
|
22
|
Sparvoli D, Delabre J, Penarete‐Vargas DM, Kumar Mageswaran S, Tsypin LM, Heckendorn J, Theveny L, Maynadier M, Mendonça Cova M, Berry‐Sterkers L, Guérin A, Dubremetz J, Urbach S, Striepen B, Turkewitz AP, Chang Y, Lebrun M. An apical membrane complex for triggering rhoptry exocytosis and invasion in Toxoplasma. EMBO J 2022; 41:e111158. [PMID: 36245278 PMCID: PMC9670195 DOI: 10.15252/embj.2022111158] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 09/13/2022] [Accepted: 09/19/2022] [Indexed: 01/13/2023] Open
Abstract
Apicomplexan parasites possess secretory organelles called rhoptries that undergo regulated exocytosis upon contact with the host. This process is essential for the parasitic lifestyle of these pathogens and relies on an exocytic machinery sharing structural features and molecular components with free-living ciliates. However, how the parasites coordinate exocytosis with host interaction is unknown. Here, we performed a Tetrahymena-based transcriptomic screen to uncover novel exocytic factors in Ciliata and conserved in Apicomplexa. We identified membrane-bound proteins, named CRMPs, forming part of a large complex essential for rhoptry secretion and invasion in Toxoplasma. Using cutting-edge imaging tools, including expansion microscopy and cryo-electron tomography, we show that, unlike previously described rhoptry exocytic factors, TgCRMPs are not required for the assembly of the rhoptry secretion machinery and only transiently associate with the exocytic site-prior to the invasion. CRMPs and their partners contain putative host cell-binding domains, and CRMPa shares similarities with GPCR proteins. Collectively our data imply that the CRMP complex acts as a host-molecular sensor to ensure that rhoptry exocytosis occurs when the parasite contacts the host cell.
Collapse
Affiliation(s)
- Daniela Sparvoli
- Laboratory of Pathogen Host InteractionsUMR 5235 CNRS, Université de MontpellierMontpellierFrance
| | - Jason Delabre
- Laboratory of Pathogen Host InteractionsUMR 5235 CNRS, Université de MontpellierMontpellierFrance
| | | | - Shrawan Kumar Mageswaran
- Department of Biochemistry and Biophysics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Lev M Tsypin
- Department of Molecular Genetics and Cell BiologyUniversity of ChicagoChicagoILUSA
- Present address:
Division of Biology and Biological EngineeringCalifornia Institute of TechnologyPasadenaCAUSA
| | - Justine Heckendorn
- Laboratory of Pathogen Host InteractionsUMR 5235 CNRS, Université de MontpellierMontpellierFrance
| | - Liam Theveny
- Department of Biochemistry and Biophysics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Marjorie Maynadier
- Laboratory of Pathogen Host InteractionsUMR 5235 CNRS, Université de MontpellierMontpellierFrance
| | - Marta Mendonça Cova
- Laboratory of Pathogen Host InteractionsUMR 5235 CNRS, Université de MontpellierMontpellierFrance
| | - Laurence Berry‐Sterkers
- Laboratory of Pathogen Host InteractionsUMR 5235 CNRS, Université de MontpellierMontpellierFrance
| | - Amandine Guérin
- Department of Pathobiology, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Jean‐François Dubremetz
- Laboratory of Pathogen Host InteractionsUMR 5235 CNRS, Université de MontpellierMontpellierFrance
| | - Serge Urbach
- IGFUniversité de Montpellier, CNRS, INSERMMontpellierFrance
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Aaron P Turkewitz
- Department of Molecular Genetics and Cell BiologyUniversity of ChicagoChicagoILUSA
| | - Yi‐Wei Chang
- Department of Biochemistry and Biophysics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Maryse Lebrun
- Laboratory of Pathogen Host InteractionsUMR 5235 CNRS, Université de MontpellierMontpellierFrance
| |
Collapse
|
23
|
Martinez M, Chen WD, Cova MM, Molnár P, Mageswaran SK, Guérin A, John ARO, Lebrun M, Chang YW. Rhoptry secretion system structure and priming in Plasmodium falciparum revealed using in situ cryo-electron tomography. Nat Microbiol 2022; 7:1230-1238. [PMID: 35817892 PMCID: PMC7613239 DOI: 10.1038/s41564-022-01171-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 06/07/2022] [Indexed: 01/13/2023]
Abstract
Apicomplexan parasites secrete contents of the rhoptries, club-shaped organelles in the apical region, into host cells to permit their invasion and establishment of infection. The rhoptry secretory apparatus (RSA), which is critical for rhoptry secretion, was recently discovered in Toxoplasma and Cryptosporidium. It is unknown whether a similar molecular machinery exists in the malaria parasite Plasmodium. In this study, we use in situ cryo-electron tomography to investigate the rhoptry secretion system in P. falciparum merozoites. We identify the presence of an RSA at the cell apex and a morphologically distinct apical vesicle docking the tips of the two rhoptries to the RSA. We also discover two additional rhoptry organizations that lack the apical vesicle. Using subtomogram averaging, we reveal different conformations of the RSA structure corresponding to different rhoptry organizations. Our results highlight previously unknown steps in the process of rhoptry secretion and indicate a regulatory role for the conserved apical vesicle in host invasion by apicomplexan parasites.
Collapse
Affiliation(s)
- Matthew Martinez
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - William David Chen
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Petra Molnár
- Department of Pediatrics, Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Shrawan Kumar Mageswaran
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amandine Guérin
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Audrey R Odom John
- Department of Pediatrics, Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Maryse Lebrun
- LPHI, UMR 5235 CNRS, Université de Montpellier, Montpellier, France
| | - Yi-Wei Chang
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
24
|
Wagner MP, Formaglio P, Gorgette O, Dziekan JM, Huon C, Berneburg I, Rahlfs S, Barale JC, Feinstein SI, Fisher AB, Ménard D, Bozdech Z, Amino R, Touqui L, Chitnis CE. Human peroxiredoxin 6 is essential for malaria parasites and provides a host-based drug target. Cell Rep 2022; 39:110923. [PMID: 35705035 DOI: 10.1016/j.celrep.2022.110923] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/30/2022] [Accepted: 05/17/2022] [Indexed: 11/30/2022] Open
Abstract
The uptake and digestion of host hemoglobin by malaria parasites during blood-stage growth leads to significant oxidative damage of membrane lipids. Repair of lipid peroxidation damage is crucial for parasite survival. Here, we demonstrate that Plasmodium falciparum imports a host antioxidant enzyme, peroxiredoxin 6 (PRDX6), during hemoglobin uptake from the red blood cell cytosol. PRDX6 is a lipid-peroxidation repair enzyme with phospholipase A2 (PLA2) activity. Inhibition of PRDX6 with a PLA2 inhibitor, Darapladib, increases lipid-peroxidation damage in the parasite and disrupts transport of hemoglobin-containing vesicles to the food vacuole, causing parasite death. Furthermore, inhibition of PRDX6 synergistically reduces the survival of artemisinin-resistant parasites following co-treatment of parasite cultures with artemisinin and Darapladib. Thus, PRDX6 is a host-derived drug target for development of antimalarial drugs that could help overcome artemisinin resistance.
Collapse
Affiliation(s)
- Matthias Paulus Wagner
- Institut Pasteur, Université de Paris, Malaria Parasite Biology and Vaccines Unit, Paris, France
| | - Pauline Formaglio
- Institut Pasteur, Université de Paris, Malaria Infection and Immunity Unit, Paris, France
| | - Olivier Gorgette
- Institut Pasteur, Department of Cell Biology and Infection, Centre for Innovation and Technological Research, Ultrastructural Bioimaging Unit, Paris, France
| | - Jerzy Michal Dziekan
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Christèle Huon
- Institut Pasteur, Université de Paris, Malaria Parasite Biology and Vaccines Unit, Paris, France
| | - Isabell Berneburg
- Biochemistry and Molecular Biology, Interdisciplinary Research Centre, Justus Liebig University Giessen, Giessen, Germany
| | - Stefan Rahlfs
- Biochemistry and Molecular Biology, Interdisciplinary Research Centre, Justus Liebig University Giessen, Giessen, Germany
| | - Jean-Christophe Barale
- Institut Pasteur, Université de Paris, CNRS UMR 3528, Structural Microbiology Unit, Paris, France; Institut Pasteur, Pasteur International Unit, Pasteur International Network, Malaria Translational Research Unit, Phnom Penh, Cambodia and Paris, France
| | | | - Aron B Fisher
- Peroxitech, Inc., Philadelphia, PA, USA; Institute for Environmental Medicine, Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Didier Ménard
- Institut Pasteur, Université de Paris, INSERM U1201, Malaria Genetics and Resistance Unit, Paris, France; Dynamics of Host-Pathogen Interactions, EA 7292, IPPTS, Strasbourg University, Strasbourg, France
| | - Zbynek Bozdech
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Rogerio Amino
- Institut Pasteur, Université de Paris, Malaria Infection and Immunity Unit, Paris, France
| | - Lhousseine Touqui
- Cystic Fibrosis, Physiopathology and Phenogenomics, INSERM Unit 938, Saint-Antoine, Paris, France; Institut Pasteur, Université de Paris, Laboratory of Cystic Fibrosis and Chronic Bronchopathies, Paris, France
| | - Chetan E Chitnis
- Institut Pasteur, Université de Paris, Malaria Parasite Biology and Vaccines Unit, Paris, France.
| |
Collapse
|
25
|
Zhang X, Wei H, Zhang Y, Zhao Y, Wang L, Hu Y, Nguitragool W, Sattabongkot J, Adams J, Cui L, Cao Y, Wang Q. Genetic diversity of Plasmodium vivax reticulocyte binding protein 2b in global parasite populations. Parasit Vectors 2022; 15:205. [PMID: 35698238 PMCID: PMC9191549 DOI: 10.1186/s13071-022-05296-6] [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: 10/27/2021] [Accepted: 04/23/2022] [Indexed: 11/19/2022] Open
Abstract
Background Plasmodium vivax reticulocyte binding protein 2b (PvRBP2b) plays a critical role in parasite invasion of reticulocytes by binding the transferrin receptor 1. PvRBP2b is a vaccine candidate based on the negative correlation between antibody titers against PvRBP2b recombinant proteins and parasitemia and risk of vivax malaria. The aim of this study was to analyze the genetic diversity of the PvRBP2b gene in the global P. vivax populations. Methods Near full-length PvRBP2b nucleotide sequences (190–8349 bp) were obtained from 88 P. vivax isolates collected from the China–Myanmar border (n = 44) and Thailand (n = 44). An additional 224 PvRBP2b sequences were retrieved from genome sequences from parasite populations worldwide. The genetic diversity, neutral selection, haplotype distribution and genetic differentiation of PvRBP2b were examined. Results The genetic diversity of PvRBP2b was distributed unevenly, with peak diversity found in the reticulocyte binding region in the N-terminus. Neutrality analysis suggested that this region is subjected to balancing selection or population bottlenecks. Several amino acid variants were found in all or nearly all P. vivax endemic regions. However, the critical residues responsible for reticulocyte binding were highly conserved. There was substantial population differentiation according to the geographical separation. The distribution of haplotypes in the reticulocyte binding region varied among regions; even the two major haplotypes Hap_6 and Hap_8 were found in only five populations. Conclusions Our data show considerable genetic variations of PvRBPb in global parasite populations. The geographic divergence may pose a challenge to PvRBP2b-based vaccine development. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13071-022-05296-6.
Collapse
Affiliation(s)
- Xuexing Zhang
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China
| | - Haichao Wei
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China.,Department of Blood Transfusion Medicine, General Hospital of Northern Theater Command, Shenyang, 110015, Liaoning, China
| | - Yangminghui Zhang
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China
| | - Yan Zhao
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China
| | - Lin Wang
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China.,Department of Blood Transfusion, Yantaishan Hospital, Yantai, 264000, Shandong, China
| | - Yubing Hu
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China.,Central Laboratory, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Wang Nguitragool
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - John Adams
- College of Public Health, Global Health Infectious Disease Research (GHIDR) Program, Tampa, FL, USA
| | - Liwang Cui
- College of Public Health, Global Health Infectious Disease Research (GHIDR) Program, Tampa, FL, USA.,Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Suite 304, Tampa, FL, 33612, USA
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China.
| | - Qinghui Wang
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
26
|
Cova MM, Lamarque MH, Lebrun M. How Apicomplexa Parasites Secrete and Build Their Invasion Machinery. Annu Rev Microbiol 2022; 76:619-640. [PMID: 35671531 DOI: 10.1146/annurev-micro-041320-021425] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Apicomplexa are obligatory intracellular parasites that sense and actively invade host cells. Invasion is a conserved process that relies on the timely and spatially controlled exocytosis of unique specialized secretory organelles termed micronemes and rhoptries. Microneme exocytosis starts first and likely controls the intricate mechanism of rhoptry secretion. To assemble the invasion machinery, micronemal proteins-associated with the surface of the parasite-interact and form complexes with rhoptry proteins, which in turn are targeted into the host cell. This review covers the molecular advances regarding microneme and rhoptry exocytosis and focuses on how the proteins discharged from these two compartments work in synergy to drive a successful invasion event. Particular emphasis is given to the structure and molecular components of the rhoptry secretion apparatus, and to the current conceptual framework of rhoptry exocytosis that may constitute an unconventional eukaryotic secretory machinery closely related to the one described in ciliates. Expected final online publication date for the Annual Review of Microbiology, Volume 76 is September 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Marta Mendonça Cova
- Laboratory of Pathogen Host Interactions (LPHI), CNRS, University of Montpellier, Montpellier, France;
| | - Mauld H Lamarque
- Laboratory of Pathogen Host Interactions (LPHI), CNRS, University of Montpellier, Montpellier, France;
| | - Maryse Lebrun
- Laboratory of Pathogen Host Interactions (LPHI), CNRS, University of Montpellier, Montpellier, France;
| |
Collapse
|
27
|
Molina-Franky J, Patarroyo ME, Kalkum M, Patarroyo MA. The Cellular and Molecular Interaction Between Erythrocytes and Plasmodium falciparum Merozoites. Front Cell Infect Microbiol 2022; 12:816574. [PMID: 35433504 PMCID: PMC9008539 DOI: 10.3389/fcimb.2022.816574] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Plasmodium falciparum is the most lethal human malaria parasite, partly due to its genetic variability and ability to use multiple invasion routes via its binding to host cell surface receptors. The parasite extensively modifies infected red blood cell architecture to promote its survival which leads to increased cell membrane rigidity, adhesiveness and permeability. Merozoites are initially released from infected hepatocytes and efficiently enter red blood cells in a well-orchestrated process that involves specific interactions between parasite ligands and erythrocyte receptors; symptoms of the disease occur during the life-cycle’s blood stage due to capillary blockage and massive erythrocyte lysis. Several studies have focused on elucidating molecular merozoite/erythrocyte interactions and host cell modifications; however, further in-depth analysis is required for understanding the parasite’s biology and thus provide the fundamental tools for developing prophylactic or therapeutic alternatives to mitigate or eliminate Plasmodium falciparum-related malaria. This review focuses on the cellular and molecular events during Plasmodium falciparum merozoite invasion of red blood cells and the alterations that occur in an erythrocyte once it has become infected.
Collapse
Affiliation(s)
- Jessica Molina-Franky
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
- Department of Immunology and Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA, United States
- PhD Programme in Biotechnology, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Manuel Elkin Patarroyo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
- Health Sciences Division, Universidad Santo Tomás, Bogotá, Colombia
- Faculty of Medicine, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Markus Kalkum
- Department of Immunology and Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA, United States
- *Correspondence: Markus Kalkum, ; Manuel Alfonso Patarroyo,
| | - Manuel Alfonso Patarroyo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
- Health Sciences Division, Universidad Santo Tomás, Bogotá, Colombia
- Faculty of Medicine, Universidad Nacional de Colombia, Bogotá, Colombia
- *Correspondence: Markus Kalkum, ; Manuel Alfonso Patarroyo,
| |
Collapse
|
28
|
Abstract
Plasmodium malaria parasites use a unique substrate-dependent locomotion, termed gliding motility, to migrate through tissues and invade cells. Previously, it was thought that the small labile invasive stages that invade erythrocytes, merozoites, use this motility solely to penetrate target erythrocytes. Here we reveal that merozoites use gliding motility for translocation across host cells prior to invasion. This forms an important preinvasion step that is powered by a conserved actomyosin motor and is regulated by a complex signaling pathway. This work broadens our understanding of the role of gliding motility and invasion in the blood and will have a significant impact on our understanding of blood stage host–pathogen interactions and parasite biology, with implications for interventions targeting erythrocyte invasion. Plasmodium malaria parasites are obligate intracellular protozoans that use a unique form of locomotion, termed gliding motility, to move through host tissues and invade cells. The process is substrate dependent and powered by an actomyosin motor that drives the posterior translocation of extracellular adhesins which, in turn, propel the parasite forward. Gliding motility is essential for tissue translocation in the sporozoite and ookinete stages; however, the short-lived erythrocyte-invading merozoite stage has never been observed to undergo gliding movement. Here we show Plasmodium merozoites possess the ability to undergo gliding motility in vitro and that this mechanism is likely an important precursor step for successful parasite invasion. We demonstrate that two human infective species, Plasmodium falciparum and Plasmodium knowlesi, have distinct merozoite motility profiles which may reflect distinct invasion strategies. Additionally, we develop and validate a higher throughput assay to evaluate the effects of genetic and pharmacological perturbations on both the molecular motor and the complex signaling cascade that regulates motility in merozoites. The discovery of merozoite motility provides a model to study the glideosome and adds a dimension for work aiming to develop treatments targeting the blood stage invasion pathways.
Collapse
|
29
|
Pereira PHS, Garcia CRS. Evidence of G-Protein-Coupled Receptors (GPCR) in the Parasitic Protozoa Plasmodium falciparum-Sensing the Host Environment and Coupling within Its Molecular Signaling Toolkit. Int J Mol Sci 2021; 22:12381. [PMID: 34830263 PMCID: PMC8620569 DOI: 10.3390/ijms222212381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 11/12/2021] [Indexed: 12/25/2022] Open
Abstract
Throughout evolution, the need for single-celled organisms to associate and form a single cluster of cells has had several evolutionary advantages. In complex, multicellular organisms, each tissue or organ has a specialty and function that make life together possible, and the organism as a whole needs to act in balance and adapt to changes in the environment. Sensory organs are essential for connecting external stimuli into a biological response, through the senses: sight, smell, taste, hearing, and touch. The G-protein-coupled receptors (GPCRs) are responsible for many of these senses and therefore play a key role in the perception of the cells' external environment, enabling interaction and coordinated development between each cell of a multicellular organism. The malaria-causing protozoan parasite, Plasmodium falciparum, has a complex life cycle that is extremely dependent on a finely regulated cellular signaling machinery. In this review, we summarize strong evidence and the main candidates of GPCRs in protozoan parasites. Interestingly, one of these GPCRs is a sensor for K+ shift in Plasmodium falciparum, PfSR25. Studying this family of proteins in P. falciparum could have a significant impact, both on understanding the history of the evolution of GPCRs and on finding new targets for antimalarials.
Collapse
Affiliation(s)
| | - Celia R. S. Garcia
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo—USP, São Paulo 05508-900, Brazil;
| |
Collapse
|
30
|
Calcium signaling in intracellular protist parasites. Curr Opin Microbiol 2021; 64:33-40. [PMID: 34571430 DOI: 10.1016/j.mib.2021.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/28/2021] [Accepted: 09/07/2021] [Indexed: 11/21/2022]
Abstract
Calcium ion (Ca2+) signaling is one of the most frequently employed mechanisms of signal transduction by eukaryotic cells, and starts with either Ca2+ release from intracellular stores or Ca2+ entry through the plasma membrane. In intracellular protist parasites Ca2+ signaling initiates a sequence of events that may facilitate their invasion of host cells, respond to environmental changes within the host, or regulate the function of their intracellular organelles. In this review we examine recent findings in Ca2+ signaling in two groups of intracellular protist parasites that have been studied in more detail, the apicomplexan and the trypanosomatid parasites.
Collapse
|
31
|
Sharma M, Choudhury H, Roy R, Michaels SA, Ojo KK, Bansal A. CDPKs: The critical decoders of calcium signal at various stages of malaria parasite development. Comput Struct Biotechnol J 2021; 19:5092-5107. [PMID: 34589185 PMCID: PMC8453137 DOI: 10.1016/j.csbj.2021.08.054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 08/31/2021] [Accepted: 08/31/2021] [Indexed: 12/13/2022] Open
Abstract
Calcium ions are used as important signals during various physiological processes. In malaria parasites, Plasmodium spp., calcium dependent protein kinases (CDPKs) have acquired the unique ability to sense and transduce calcium signals at various critical steps during the lifecycle, either through phosphorylation of downstream substrates or mediating formation of high molecular weight protein complexes. Calcium signaling cascades establish important crosstalk events with signaling pathways mediated by other secondary messengers such as cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). CDPKs play critical roles at various important physiological steps during parasite development in vertebrates and mosquitoes. They are also important for transmission of the parasite between the two hosts. Combined with the fact that CDPKs are not present in humans, they continue to be pursued as important targets for development of anti-malarial drugs.
Collapse
Affiliation(s)
- Manish Sharma
- Molecular Parasitology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Himashree Choudhury
- Molecular Parasitology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rajarshi Roy
- Molecular Parasitology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Samantha A. Michaels
- Center for Emerging and Re-emerging Infectious Diseases, Division of Allergy & Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA 98109 USA
| | - Kayode K. Ojo
- Center for Emerging and Re-emerging Infectious Diseases, Division of Allergy & Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA 98109 USA
| | - Abhisheka Bansal
- Molecular Parasitology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| |
Collapse
|
32
|
Miao J, Wang C, Lucky AB, Liang X, Min H, Adapa SR, Jiang R, Kim K, Cui L. A unique GCN5 histone acetyltransferase complex controls erythrocyte invasion and virulence in the malaria parasite Plasmodium falciparum. PLoS Pathog 2021; 17:e1009351. [PMID: 34403450 PMCID: PMC8396726 DOI: 10.1371/journal.ppat.1009351] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 08/27/2021] [Accepted: 07/21/2021] [Indexed: 12/14/2022] Open
Abstract
The histone acetyltransferase GCN5-associated SAGA complex is evolutionarily conserved from yeast to human and functions as a general transcription co-activator in global gene regulation. In this study, we identified a divergent GCN5 complex in Plasmodium falciparum, which contains two plant homeodomain (PHD) proteins (PfPHD1 and PfPHD2) and a plant apetela2 (AP2)-domain transcription factor (PfAP2-LT). To dissect the functions of the PfGCN5 complex, we generated parasite lines with either the bromodomain in PfGCN5 or the PHD domain in PfPHD1 deleted. The two deletion mutants closely phenocopied each other, exhibiting significantly reduced merozoite invasion of erythrocytes and elevated sexual conversion. These domain deletions caused dramatic decreases not only in histone H3K9 acetylation but also in H3K4 trimethylation, indicating synergistic crosstalk between the two euchromatin marks. Domain deletion in either PfGCN5 or PfPHD1 profoundly disturbed the global transcription pattern, causing altered expression of more than 60% of the genes. At the schizont stage, these domain deletions were linked to specific down-regulation of merozoite genes involved in erythrocyte invasion, many of which contain the AP2-LT binding motif and are also regulated by AP2-I and BDP1, suggesting targeted recruitment of the PfGCN5 complex to the invasion genes by these specific factors. Conversely, at the ring stage, PfGCN5 or PfPHD1 domain deletions disrupted the mutually exclusive expression pattern of the entire var gene family, which encodes the virulent factor PfEMP1. Correlation analysis between the chromatin state and alteration of gene expression demonstrated that up- and down-regulated genes in these mutants are highly correlated with the silent and active chromatin states in the wild-type parasite, respectively. Collectively, the PfGCN5 complex represents a novel HAT complex with a unique subunit composition including an AP2 transcription factor, which signifies a new paradigm for targeting the co-activator complex to regulate general and parasite-specific cellular processes in this low-branching parasitic protist. Epigenetic regulation of gene expression plays essential roles in orchestrating the general and parasite-specific cellular pathways in the malaria parasite Plasmodium falciparum. To better understand the epigenetic mechanisms in this parasite, we characterized the histone acetyltransferase GCN5-mediated transcription regulation during intraerythrocytic development of the parasite. Using tandem affinity purification and proteomic characterization, we identified that the PfGCN5-associated complex contains nine core components, including two PHD domain proteins (PfPHD1 and PfPHD2) and an AP2-domain transcription factor, which is divergent from the canonical GCN5 complexes evolutionarily conserved from yeast to human. To understand the functions of the PfGCN5 complex, we performed domain deletions in two subunits of this complex, PfGCN5 and PfPHD1. We found that the two deletion mutants displayed very similar growth phenotypes, including significantly reduced merozoite invasion rates and elevated sexual conversion. These two mutants were associated with dramatic decreases in histone H3K9 acetylation and H3K4 trimethylation, which led to global changes in chromatin states and gene expression. Consistent with the phenotypes, genes significantly affected by the PfGCN5 and PfPHD1 gene disruption include those participating in parasite-specific pathways such as invasion, virulence, and sexual development. In conclusion, this study presents a new model of the PfGCN5 complex for targeting the co-activator complex to regulate general and parasite-specific cellular processes in this low-branching parasitic protist.
Collapse
Affiliation(s)
- Jun Miao
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
- * E-mail: (JM); (LC)
| | - Chengqi Wang
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Amuza Byaruhanga Lucky
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Xiaoying Liang
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Hui Min
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Swamy Rakesh Adapa
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Rays Jiang
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Kami Kim
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
- * E-mail: (JM); (LC)
| |
Collapse
|
33
|
Mageswaran SK, Guérin A, Theveny LM, Chen WD, Martinez M, Lebrun M, Striepen B, Chang YW. In situ ultrastructures of two evolutionarily distant apicomplexan rhoptry secretion systems. Nat Commun 2021; 12:4983. [PMID: 34404783 PMCID: PMC8371170 DOI: 10.1038/s41467-021-25309-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 08/03/2021] [Indexed: 12/03/2022] Open
Abstract
Parasites of the phylum Apicomplexa cause important diseases including malaria, cryptosporidiosis and toxoplasmosis. These intracellular pathogens inject the contents of an essential organelle, the rhoptry, into host cells to facilitate invasion and infection. However, the structure and mechanism of this eukaryotic secretion system remain elusive. Here, using cryo-electron tomography and subtomogram averaging, we report the conserved architecture of the rhoptry secretion system in the invasive stages of two evolutionarily distant apicomplexans, Cryptosporidium parvum and Toxoplasma gondii. In both species, we identify helical filaments, which appear to shape and compartmentalize the rhoptries, and an apical vesicle (AV), which facilitates docking of the rhoptry tip at the parasite's apical region with the help of an elaborate ultrastructure named the rhoptry secretory apparatus (RSA); the RSA anchors the AV at the parasite plasma membrane. Depletion of T. gondii Nd9, a protein required for rhoptry secretion, disrupts the RSA ultrastructure and AV-anchoring. Moreover, T. gondii contains a line of AV-like vesicles, which interact with a pair of microtubules and accumulate towards the AV, leading to a working model for AV-reloading and discharging of multiple rhoptries. Together, our analyses provide an ultrastructural framework to understand how these important parasites deliver effectors into host cells.
Collapse
Affiliation(s)
- Shrawan Kumar Mageswaran
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amandine Guérin
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Liam M Theveny
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - William David Chen
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew Martinez
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maryse Lebrun
- LPHI, UMR 5235 CNRS, Université de Montpellier, Montpellier, France
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yi-Wei Chang
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
34
|
Ishizaki T, Asada M, Hakimi H, Chaiyawong N, Kegawa Y, Yahata K, Kaneko O. cAMP-dependent protein kinase regulates secretion of apical membrane antigen 1 (AMA1) in Plasmodium yoelii. Parasitol Int 2021; 85:102435. [PMID: 34390881 DOI: 10.1016/j.parint.2021.102435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/06/2021] [Accepted: 08/06/2021] [Indexed: 12/18/2022]
Abstract
Malaria remains a heavy global burden on human health, and it is important to understand the molecular and cellular biology of the parasite to find targets for drug and vaccine development. The mouse malaria model is an essential tool to characterize the function of identified molecules; however, robust technologies for targeted gene deletions are still poorly developed for the widely used rodent malaria parasite, Plasmodium yoelii. To overcome this problem, we established a DiCre-loxP inducible knockout (iKO) system in P. yoelii, which showed more than 80% excision efficacy of the target locus and more than 90% reduction of locus transcripts 24 h (one cell cycle) after RAP administration. Using this developed system, cAMP-dependent protein kinase (PKAc) was inducibly disrupted and the phenotypes of the resulting PKAc-iKO parasites were analyzed. We found that PKAc-iKO parasites showed severe growth and erythrocyte invasion defects. We also found that disruption of PKAc impaired the secretion of AMA1 in P. yoelii, in contrast to a report showing no role of PKAc in AMA1 secretion in P. falciparum. This discrepancy may be related to the difference in the timing of AMA1 distribution to the merozoite surface, which occurs just after egress for P. falciparum, but after several minutes for P. yoelii. Secretions of PyEBL, Py235, and RON2 were not affected by the disruption of PKAc in P. yoelii. PyRON2 was already secreted to the merozoite surface immediately after merozoite egress, which is inconsistent with the current model that RON2 is injected into the erythrocyte cytosol. Further investigations are required to understand the role of RON2 exposed on the merozoite surface.
Collapse
Affiliation(s)
- Takahiro Ishizaki
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate school of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Laboratory for Molecular Infection Medicine Sweden, Department of Molecular Biology, Umeå University, Umeå 901 87, Sweden
| | - Masahito Asada
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate school of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Nishi 2-11, Obihiro, Hokkaido 080-0834, Japan.
| | - Hassan Hakimi
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College station, TX 77843, USA.
| | - Nattawat Chaiyawong
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate school of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Yuto Kegawa
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate school of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Section on Integrative Biophysics, Eunice Kennedy Shriver National Institutes of Child Health and Human Development, National Institute of Health, 9000 Rockville Pike, Bethesda, Mary land 20892, USA
| | - Kazuhide Yahata
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan.
| | - Osamu Kaneko
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate school of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan.
| |
Collapse
|
35
|
de Oliveira LS, Alborghetti MR, Carneiro RG, Bastos IMD, Amino R, Grellier P, Charneau S. Calcium in the Backstage of Malaria Parasite Biology. Front Cell Infect Microbiol 2021; 11:708834. [PMID: 34395314 PMCID: PMC8355824 DOI: 10.3389/fcimb.2021.708834] [Citation(s) in RCA: 6] [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: 05/12/2021] [Accepted: 07/14/2021] [Indexed: 12/26/2022] Open
Abstract
The calcium ion (Ca2+) is a ubiquitous second messenger involved in key biological processes in prokaryotes and eukaryotes. In Plasmodium species, Ca2+ signaling plays a central role in the parasite life cycle. It has been associated with parasite development, fertilization, locomotion, and host cell infection. Despite the lack of a canonical inositol-1,4,5-triphosphate receptor gene in the Plasmodium genome, pharmacological evidence indicates that inositol-1,4,5-triphosphate triggers Ca2+ mobilization from the endoplasmic reticulum. Other structures such as acidocalcisomes, food vacuole and mitochondria are proposed to act as supplementary intracellular Ca2+ reservoirs. Several Ca2+-binding proteins (CaBPs) trigger downstream signaling. Other proteins with no EF-hand motifs, but apparently involved with CaBPs, are depicted as playing an important role in the erythrocyte invasion and egress. It is also proposed that a cross-talk among kinases, which are not members of the family of Ca2+-dependent protein kinases, such as protein kinases G, A and B, play additional roles mediated indirectly by Ca2+ regulation. This statement may be extended for proteins directly related to invasion or egress, such as SUB1, ERC, IMC1I, IMC1g, GAP45 and EBA175. In this review, we update our understanding of aspects of Ca2+-mediated signaling correlated to the developmental stages of the malaria parasite life cycle.
Collapse
Affiliation(s)
- Lucas Silva de Oliveira
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
- UMR 7245 MCAM, Molécules de Communication et Adaptation des Micro-organismes, Muséum National d’Histoire Naturelle, CNRS, Équipe Parasites et Protistes Libres, Paris, France
| | - Marcos Rodrigo Alborghetti
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| | - Renata Garcia Carneiro
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Izabela Marques Dourado Bastos
- Laboratory of Host-Pathogen Interaction, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Rogerio Amino
- Unité Infection et Immunité Paludéennes, Institut Pasteur, Paris, France
| | - Philippe Grellier
- UMR 7245 MCAM, Molécules de Communication et Adaptation des Micro-organismes, Muséum National d’Histoire Naturelle, CNRS, Équipe Parasites et Protistes Libres, Paris, France
| | - Sébastien Charneau
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| |
Collapse
|
36
|
Pereira PHS, Borges-Pereira L, Garcia CRS. Evidences of G Coupled-Protein Receptor (GPCR) Signaling in the human Malaria Parasite Plasmodium falciparum for Sensing its Microenvironment and the Role of Purinergic Signaling in Malaria Parasites. Curr Top Med Chem 2021; 21:171-180. [PMID: 32851963 DOI: 10.2174/1568026620666200826122716] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/15/2020] [Accepted: 07/20/2020] [Indexed: 11/22/2022]
Abstract
The nucleotides were discovered in the early 19th century and a few years later, the role of such molecules in energy metabolism and cell survival was postulated. In 1972, a pioneer work by Burnstock and colleagues suggested that ATP could also work as a neurotransmitter, which was known as the "purinergic hypothesis". The idea of ATP working as a signaling molecule faced initial resistance until the discovery of the receptors for ATP and other nucleotides, called purinergic receptors. Among the purinergic receptors, the P2Y family is of great importance because it comprises of G proteincoupled receptors (GPCRs). GPCRs are widespread among different organisms. These receptors work in the cells' ability to sense the external environment, which involves: to sense a dangerous situation or detect a pheromone through smell; the taste of food that should not be eaten; response to hormones that alter metabolism according to the body's need; or even transform light into an electrical stimulus to generate vision. Advances in understanding the mechanism of action of GPCRs shed light on increasingly promising treatments for diseases that have hitherto remained incurable, or the possibility of abolishing side effects from therapies widely used today.
Collapse
Affiliation(s)
- Pedro H S Pereira
- Department of Clinical and Toxicological Analyses, University of Sao Paulo, Sao Paulo, Brazil
| | - Lucas Borges-Pereira
- Department of Clinical and Toxicological Analyses, University of Sao Paulo, Sao Paulo, Brazil
| | - Célia R S Garcia
- Department of Clinical and Toxicological Analyses, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
37
|
Zhu J, Wang Y, Cao Y, Shen J, Yu L. Diverse Roles of TgMIC1/4/6 in the Toxoplasma Infection. Front Microbiol 2021; 12:666506. [PMID: 34220751 PMCID: PMC8247436 DOI: 10.3389/fmicb.2021.666506] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/25/2021] [Indexed: 11/23/2022] Open
Abstract
Toxoplasma gondii microneme is a specialized secretory organelle that discharges its contents at the apical tip of this apicomplexan parasite in a sequential and regulated manner. Increasing number of studies on microneme proteins (MICs) have shown them as a predominant and important role in host cell attachment, invasion, motility and pathogenesis. In this review, we summarize the research advances in one of the most important MICs complexes, TgMIC1/4/6, which will contribute to improve the understanding of the molecular mechanism of T. gondii infection and provide a theoretical basis for the effective control against T. gondii.
Collapse
Affiliation(s)
- Jinjin Zhu
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yang Wang
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yuanyuan Cao
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jilong Shen
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Li Yu
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
38
|
Lasonder E, More K, Singh S, Haidar M, Bertinetti D, Kennedy EJ, Herberg FW, Holder AA, Langsley G, Chitnis CE. cAMP-Dependent Signaling Pathways as Potential Targets for Inhibition of Plasmodium falciparum Blood Stages. Front Microbiol 2021; 12:684005. [PMID: 34108954 PMCID: PMC8183823 DOI: 10.3389/fmicb.2021.684005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/30/2021] [Indexed: 11/13/2022] Open
Abstract
We review the role of signaling pathways in regulation of the key processes of merozoite egress and red blood cell invasion by Plasmodium falciparum and, in particular, the importance of the second messengers, cAMP and Ca2+, and cyclic nucleotide dependent kinases. cAMP-dependent protein kinase (PKA) is comprised of cAMP-binding regulatory, and catalytic subunits. The less well conserved cAMP-binding pockets should make cAMP analogs attractive drug leads, but this approach is compromised by the poor membrane permeability of cyclic nucleotides. We discuss how the conserved nature of ATP-binding pockets makes ATP analogs inherently prone to off-target effects and how ATP analogs and genetic manipulation can be useful research tools to examine this. We suggest that targeting PKA interaction partners as well as substrates, or developing inhibitors based on PKA interaction sites or phosphorylation sites in PKA substrates, may provide viable alternative approaches for the development of anti-malarial drugs. Proximity of PKA to a substrate is necessary for substrate phosphorylation, but the P. falciparum genome encodes few recognizable A-kinase anchor proteins (AKAPs), suggesting the importance of PKA-regulatory subunit myristylation and membrane association in determining substrate preference. We also discuss how Pf14-3-3 assembles a phosphorylation-dependent signaling complex that includes PKA and calcium dependent protein kinase 1 (CDPK1) and how this complex may be critical for merozoite invasion, and a target to block parasite growth. We compare altered phosphorylation levels in intracellular and egressed merozoites to identify potential PKA substrates. Finally, as host PKA may have a critical role in supporting intracellular parasite development, we discuss its role at other stages of the life cycle, as well as in other apicomplexan infections. Throughout our review we propose possible new directions for the therapeutic exploitation of cAMP-PKA-signaling in malaria and other diseases caused by apicomplexan parasites.
Collapse
Affiliation(s)
- Edwin Lasonder
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, United Kingdom
| | - Kunal More
- Unité de Biologie de Plasmodium et Vaccins, Département de Parasites et Insectes Vecteurs, Institut Pasteur, Paris, France
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Malak Haidar
- Laboratoire de Biologie Comparative des Apicomplexes, Faculté de Médecine, Université Paris Descartes - Sorbonne Paris Cité, Paris, France.,INSERM U1016, CNRS UMR 8104, Cochin Institute, Paris, France
| | | | - Eileen J Kennedy
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, United States
| | | | - Anthony A Holder
- Malaria Parasitology Laboratory, Francis Crick Institute, London, United Kingdom
| | - Gordon Langsley
- Laboratoire de Biologie Comparative des Apicomplexes, Faculté de Médecine, Université Paris Descartes - Sorbonne Paris Cité, Paris, France.,INSERM U1016, CNRS UMR 8104, Cochin Institute, Paris, France
| | - Chetan E Chitnis
- Unité de Biologie de Plasmodium et Vaccins, Département de Parasites et Insectes Vecteurs, Institut Pasteur, Paris, France
| |
Collapse
|
39
|
Sparvoli D, Lebrun M. Unraveling the Elusive Rhoptry Exocytic Mechanism of Apicomplexa. Trends Parasitol 2021; 37:622-637. [PMID: 34045149 DOI: 10.1016/j.pt.2021.04.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022]
Abstract
Apicomplexan parasites are unicellular eukaryotes that invade the cells in which they proliferate. The development of genetic tools in Toxoplasma, and then in Plasmodium, in the 1990s allowed the first description of the molecular machinery used for motility and invasion, revealing a crucial role for two different secretory organelles, micronemes and rhoptries. Rhoptry proteins are injected directly into the host cytoplasm not only to promote invasion but also to manipulate host functions. Nonetheless, the injection machinery has remained mysterious, a major conundrum in the field. Here we review recent progress in uncovering structural components and proteins implicated in rhoptry exocytosis and explain how revisiting early findings and considering the evolutionary origins of Apicomplexa contributed to some of these discoveries.
Collapse
Affiliation(s)
- Daniela Sparvoli
- LPHI UMR5235, Univ Montpellier, CNRS, F-34095 Montpellier, France
| | - Maryse Lebrun
- LPHI UMR5235, Univ Montpellier, CNRS, F-34095 Montpellier, France.
| |
Collapse
|
40
|
Loubens M, Vincensini L, Fernandes P, Briquet S, Marinach C, Silvie O. Plasmodium sporozoites on the move: Switching from cell traversal to productive invasion of hepatocytes. Mol Microbiol 2021; 115:870-881. [PMID: 33191548 PMCID: PMC8247013 DOI: 10.1111/mmi.14645] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/18/2022]
Abstract
Parasites of the genus Plasmodium, the etiological agent of malaria, are transmitted through the bite of anopheline mosquitoes, which deposit sporozoites into the host skin. Sporozoites migrate through the dermis, enter the bloodstream, and rapidly traffic to the liver. They cross the liver sinusoidal barrier and traverse several hepatocytes before switching to productive invasion of a final one for replication inside a parasitophorous vacuole. Cell traversal and productive invasion are functionally independent processes that require proteins secreted from specialized secretory organelles known as micronemes. In this review, we summarize the current understanding of how sporozoites traverse through cells and productively invade hepatocytes, and discuss the role of environmental sensing in switching from a migratory to an invasive state. We propose that timely controlled secretion of distinct microneme subsets could play a key role in successful migration and infection of hepatocytes. A better understanding of these essential biological features of the Plasmodium sporozoite may contribute to the development of new strategies to fight against the very first and asymptomatic stage of malaria.
Collapse
Affiliation(s)
- Manon Loubens
- Centre d’Immunologie et des Maladies InfectieusesSorbonne Université, INSERM, CNRS, CIMI‐ParisParisFrance
| | - Laetitia Vincensini
- Centre d’Immunologie et des Maladies InfectieusesSorbonne Université, INSERM, CNRS, CIMI‐ParisParisFrance
| | - Priyanka Fernandes
- Centre d’Immunologie et des Maladies InfectieusesSorbonne Université, INSERM, CNRS, CIMI‐ParisParisFrance
| | - Sylvie Briquet
- Centre d’Immunologie et des Maladies InfectieusesSorbonne Université, INSERM, CNRS, CIMI‐ParisParisFrance
| | - Carine Marinach
- Centre d’Immunologie et des Maladies InfectieusesSorbonne Université, INSERM, CNRS, CIMI‐ParisParisFrance
| | - Olivier Silvie
- Centre d’Immunologie et des Maladies InfectieusesSorbonne Université, INSERM, CNRS, CIMI‐ParisParisFrance
| |
Collapse
|
41
|
Santos BM, Dias BKM, Nakabashi M, Garcia CRS. The Knockout for G Protein-Coupled Receptor-Like PfSR25 Increases the Susceptibility of Malaria Parasites to the Antimalarials Lumefantrine and Piperaquine but Not to Medicine for Malaria Venture Compounds. Front Microbiol 2021; 12:638869. [PMID: 33790879 PMCID: PMC8006397 DOI: 10.3389/fmicb.2021.638869] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/17/2021] [Indexed: 12/11/2022] Open
Abstract
Previously we have reported that the G protein-coupled receptor (GPCR)-like PfSR25 in Plasmodium falciparum is a potassium (K+) sensor linked to intracellular calcium signaling and that knockout parasites (PfSR25-) are more susceptible to oxidative stress and antimalarial compounds. Here, we explore the potential role of PfSR25 in susceptibility to the antimalarial compounds atovaquone, chloroquine, dihydroartemisinin, lumefantrine, mefloquine, piperaquine, primaquine, and pyrimethamine and the Medicine for Malaria Venture (MMV) compounds previously described to act on egress/invasion (MMV006429, MMV396715, MMV019127, MMV665874, MMV665878, MMV665785, and MMV66583) through comparative assays with PfSR25- and 3D7 parasite strains, using flow cytometry assays. The IC50 and IC90 results show that lumefantrine and piperaquine have greater activity on the PfSR25- parasite strain when compared to 3D7. For MMV compounds, we found no differences between the strains except for the compound MMV665831, which we used to investigate the store-operated calcium entry (SOCE) mechanism. The results suggest that PfSR25 may be involved in the mechanism of action of the antimalarials lumefantrine and piperaquine. Our data clearly show that MMV665831 does not affect calcium entry in parasites after we depleted their internal calcium pools with thapsigargin. The results demonstrated here shed light on new possibilities on the antimalarial mechanism, bringing evidence of the involvement of the GPCR-like PfSR25.
Collapse
Affiliation(s)
- Benedito M Santos
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Bárbara K M Dias
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Myna Nakabashi
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Celia R S Garcia
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
42
|
Aquilini E, Cova MM, Mageswaran SK, Dos Santos Pacheco N, Sparvoli D, Penarete-Vargas DM, Najm R, Graindorge A, Suarez C, Maynadier M, Berry-Sterkers L, Urbach S, Fahy PR, Guérin AN, Striepen B, Dubremetz JF, Chang YW, Turkewitz AP, Lebrun M. An Alveolata secretory machinery adapted to parasite host cell invasion. Nat Microbiol 2021; 6:425-434. [PMID: 33495622 PMCID: PMC8886610 DOI: 10.1038/s41564-020-00854-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 12/14/2020] [Indexed: 01/29/2023]
Abstract
Apicomplexa are unicellular eukaryotes and obligate intracellular parasites, including Plasmodium (the causative agent of malaria) and Toxoplasma (one of the most widespread zoonotic pathogens). Rhoptries, one of their specialized secretory organelles, undergo regulated exocytosis during invasion1. Rhoptry proteins are injected directly into the host cell to support invasion and subversion of host immune function2. The mechanism by which they are discharged is unclear and appears distinct from those in bacteria, yeast, animals and plants. Here, we show that rhoptry secretion in Apicomplexa shares structural and genetic elements with the exocytic machinery of ciliates, their free-living relatives. Rhoptry exocytosis depends on intramembranous particles in the shape of a rosette embedded into the plasma membrane of the parasite apex. Formation of this rosette requires multiple non-discharge (Nd) proteins conserved and restricted to Ciliata, Dinoflagellata and Apicomplexa that together constitute the superphylum Alveolata. We identified Nd6 at the site of exocytosis in association with an apical vesicle. Sandwiched between the rosette and the tip of the rhoptry, this vesicle appears as a central element of the rhoptry secretion machine. Our results describe a conserved secretion system that was adapted to provide defence for free-living unicellular eukaryotes and host cell injection in intracellular parasites.
Collapse
Affiliation(s)
- Eleonora Aquilini
- Laboratory of Pathogen Host Interactions UMR 5235, Université de Montpellier and CNRS, Montpellier, France
| | - Marta Mendonça Cova
- Laboratory of Pathogen Host Interactions UMR 5235, Université de Montpellier and CNRS, Montpellier, France
| | - Shrawan Kumar Mageswaran
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicolas Dos Santos Pacheco
- Laboratory of Pathogen Host Interactions UMR 5235, Université de Montpellier and CNRS, Montpellier, France
| | - Daniela Sparvoli
- Laboratory of Pathogen Host Interactions UMR 5235, Université de Montpellier and CNRS, Montpellier, France
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| | | | - Rania Najm
- Laboratory of Pathogen Host Interactions UMR 5235, Université de Montpellier and CNRS, Montpellier, France
| | - Arnault Graindorge
- Laboratory of Pathogen Host Interactions UMR 5235, Université de Montpellier and CNRS, Montpellier, France
| | - Catherine Suarez
- Laboratory of Pathogen Host Interactions UMR 5235, Université de Montpellier and CNRS, Montpellier, France
| | - Marjorie Maynadier
- Laboratory of Pathogen Host Interactions UMR 5235, Université de Montpellier and CNRS, Montpellier, France
| | - Laurence Berry-Sterkers
- Laboratory of Pathogen Host Interactions UMR 5235, Université de Montpellier and CNRS, Montpellier, France
| | - Serge Urbach
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS and INSERM, Montpellier, France
| | - Pilar Ruga Fahy
- Pôle Facultaire de Microscopie Ultrastructurale, Geneva, Switzerland
| | - Amandine N Guérin
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jean-François Dubremetz
- Laboratory of Pathogen Host Interactions UMR 5235, Université de Montpellier and CNRS, Montpellier, France
| | - Yi-Wei Chang
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aaron P Turkewitz
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| | - Maryse Lebrun
- Laboratory of Pathogen Host Interactions UMR 5235, Université de Montpellier and CNRS, Montpellier, France.
| |
Collapse
|
43
|
Abundant Monovalent Ions as Environmental Signposts for Pathogens during Host Colonization. Infect Immun 2021; 89:IAI.00641-20. [PMID: 33526568 DOI: 10.1128/iai.00641-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Host colonization by a pathogen requires proper sensing and response to local environmental cues, to ensure adaptation and continued survival within the host. The ionic milieu represents a critical potential source of environmental cues, and indeed, there has been extensive study of the interplay between host and pathogen in the context of metals such as iron, zinc, and manganese, vital ions that are actively sequestered by the host. The inherent non-uniformity of the ionic milieu also extends, however, to "abundant" ions such as chloride and potassium, whose concentrations vary greatly between tissue and cellular locations, and with the immune response. Despite this, the concept of abundant ions as environmental cues and key players in host-pathogen interactions is only just emerging. Focusing on chloride and potassium, this review brings together studies across multiple bacterial and parasitic species that have begun to define both how these abundant ions are exploited as cues during host infection, and how they can be actively manipulated by pathogens during host colonization. The close links between ion homeostasis and sensing/response to different ionic signals, and the importance of studying pathogen response to cues in combination, are also discussed, while considering the fundamental insight still to be uncovered from further studies in this nascent area of inquiry.
Collapse
|
44
|
Scarpelli PH, Pecenin MF, Garcia CRS. Intracellular Ca 2+ Signaling in Protozoan Parasites: An Overview with a Focus on Mitochondria. Int J Mol Sci 2021; 22:ijms22010469. [PMID: 33466510 PMCID: PMC7796463 DOI: 10.3390/ijms22010469] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/07/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
Ca2+ signaling has been involved in controling critical cellular functions such as activation of proteases, cell death, and cell cycle control. The endoplasmatic reticulum plays a significant role in Ca2+ storage inside the cell, but mitochondria have long been recognized as a fundamental Ca2+ pool. Protozoan parasites such as Plasmodium falciparum, Toxoplasma gondii, and Trypanosoma cruzi display a Ca2+ signaling toolkit with similarities to higher eukaryotes, including the participation of mitochondria in Ca2+-dependent signaling events. This review summarizes the most recent knowledge in mitochondrial Ca2+ signaling in protozoan parasites, focusing on the mechanism involved in mitochondrial Ca2+ uptake by pathogenic protists.
Collapse
|
45
|
Uboldi AD, Wilde ML, Bader SM, Tonkin CJ. Environmental sensing and regulation of motility in Toxoplasma. Mol Microbiol 2020; 115:916-929. [PMID: 33278047 DOI: 10.1111/mmi.14661] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/27/2020] [Accepted: 12/01/2020] [Indexed: 12/28/2022]
Abstract
Toxoplasma and other apicomplexan parasites undergo a unique form of cellular locomotion referred to as "gliding motility." Gliding motility is crucial for parasite survival as it powers tissue dissemination, host cell invasion and egress. Distinct environmental cues lead to activation of gliding motility and have become a prominent focus of recent investigation. Progress has been made toward understanding what environmental cues are sensed and how these signals are transduced in order to regulate the machinery and cellular events powering gliding motility. In this review, we will discuss new findings and integrate these into our current understanding to propose a model of how environmental sensing is achieved to regulate gliding motility in Toxoplasma. Collectively, these findings also have implications for the understanding of gliding motility across Apicomplexa more broadly.
Collapse
Affiliation(s)
- Alessandro D Uboldi
- Division of Infectious Disease and Immune Defense, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,The Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Mary-Louise Wilde
- Division of Infectious Disease and Immune Defense, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,The Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Stefanie M Bader
- Division of Infectious Disease and Immune Defense, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Christopher J Tonkin
- Division of Infectious Disease and Immune Defense, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,The Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
46
|
Elsworth B, Duraisingh MT. A framework for signaling throughout the life cycle of Babesia species. Mol Microbiol 2020; 115:882-890. [PMID: 33274587 DOI: 10.1111/mmi.14650] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 01/28/2023]
Abstract
Babesia species are tick-borne intracellular parasites that infect the red blood cells of their mammalian host, leading to severe or fatal disease. Babesia spp. infect a wide range of mammalian species and cause a significant economic burden globally, predominantly through disease in cattle. Several Babesia spp. are increasingly being recognized as zoonotic pathogens of humans. Babesia spp. have complex life cycles involving multiple stages in the tick and the mammalian host. The parasite utilizes complex signaling pathways during replication, egress, and invasion in each of these stages. They must also rapidly respond to their environment when switching between the mammalian and tick stages. This review will focus on the signaling pathways and environmental stimuli that Babesia spp. utilize in the bloodstream and for transmission to the tick, with an emphasis on the role of phosphorylation- and calcium-based signaling during egress and invasion. The expanding availability of in vitro and in vivo culture systems, genomes, transcriptomes, and transgenic systems available for a range of Babesia spp. should encourage further biological and translational studies of these ubiquitous parasites.
Collapse
Affiliation(s)
- Brendan Elsworth
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Manoj T Duraisingh
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
47
|
Development of novel anti-malarial from structurally diverse library of molecules, targeting plant-like CDPK1, a multistage growth regulator of P. falciparum. Biochem J 2020; 477:1951-1970. [PMID: 32401306 DOI: 10.1042/bcj20200045] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 05/11/2020] [Accepted: 05/13/2020] [Indexed: 01/06/2023]
Abstract
Upon Plasmodium falciparum merozoites exposure to low [K+] environment in blood plasma, there is escalation of cytosolic [Ca2+] which activates Ca2+-Dependent Protein Kinase 1 (CDPK1), a signaling hub of intra-erythrocytic proliferative stages of parasite. Given its high abundance and multidimensional attributes in parasite life-cycle, this is a lucrative target for designing antimalarials. Towards this, we have virtually screened MyriaScreenII diversity collection of 10,000 drug-like molecules, which resulted in 18 compounds complementing ATP-binding pocket of CDPK1. In vitro screening for toxicity in mammalian cells revealed that these compounds are non-toxic in nature. Furthermore, SPR analysis demonstrated differential binding affinity of these compounds towards recombinantly purified CDPK1 protein. Selection of lead compound 1 was performed by evaluating their inhibitory effects on phosphorylation and ATP binding activities of CDPK1. Furthermore, in vitro biophysical evaluations by ITC and FS revealed that binding of compound 1 is driven by formation of energetically favorable non-covalent interactions, with different binding constants in presence and absence of Ca2+, and TSA authenticated stability of compound 1 bound CDPK1 complex. Finally, compound 1 strongly inhibited intra-erythrocytic growth of P. falciparum in vitro. Conceivably, we propose a novel CDPK1-selective inhibitor, step towards developing pan-CDPK kinase inhibitors, prerequisite for cross-stage anti-malarial protection.
Collapse
|
48
|
Vo KC, Günay-Esiyok Ö, Liem N, Gupta N. The protozoan parasite Toxoplasma gondii encodes a gamut of phosphodiesterases during its lytic cycle in human cells. Comput Struct Biotechnol J 2020; 18:3861-3876. [PMID: 33335684 PMCID: PMC7720076 DOI: 10.1016/j.csbj.2020.11.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 11/11/2020] [Accepted: 11/13/2020] [Indexed: 01/21/2023] Open
Abstract
Toxoplasma genome harbors at least 18 phosphodiesterases encoded by distinct genes. Most parasite PDEs lack regulatory modules and are quite divergent from their human orthologs. Acutely-infectious tachyzoite stage of T. gondii expresses 11 PDEs with varied localizations. PDE8 and PDE9 are closely-related dual-substrate specific proteins residing in the apical pole. Homology modeling of PDE8 and PDE9 reveals a conserved 3D topology and substrate pocket. PDE9 is dispensable in tachyzoites, signifying a functional redundancy with PDE8.
Cyclic nucleotide signaling is pivotal to the asexual reproduction of Toxoplasma gondii, however little do we know about the phosphodiesterase enzymes in this widespread obligate intracellular parasite. Here, we identified 18 phosphodiesterases (TgPDE1-18) in the parasite genome, most of which form apicomplexan-specific clades and lack archetypal regulatory motifs often found in mammalian PDEs. Genomic epitope-tagging in the tachyzoite stage showed the expression of 11 phosphodiesterases with diverse subcellular distributions. Notably, TgPDE8 and TgPDE9 are located in the apical plasma membrane to regulate cAMP and cGMP signaling, as suggested by their dual-substrate catalysis and structure modeling. TgPDE9 expression can be ablated with no apparent loss of growth fitness in tachyzoites. Likewise, the redundancy in protein expression, subcellular localization and predicted substrate specificity of several other PDEs indicate significant plasticity and spatial control of cyclic nucleotide signaling during the lytic cycle. Our findings shall enable a rational dissection of signaling in tachyzoites by combinatorial mutagenesis. Moreover, the phylogenetic divergence of selected Toxoplasma PDEs from human counterparts can be exploited to develop parasite-specific inhibitors and therapeutics.
Collapse
Key Words
- 3′IT, 3′-insertional tagging
- AC, adenylate cyclase
- Apicomplexa
- Bradyzoite
- COS, crossover sequence
- CRISPR, clustered regularly interspaced short palindromic repeats
- EES, entero-epithelial stages
- FPKM, fragments per kilobase of exon model per million
- GC, guanylate cyclase
- GMQE, Global Model Quality Estimation
- HFF, human foreskin fibroblast
- HXGPRT, hypoxanthine-xanthine-guanine phosphoribosyltransferase
- IMC, inner membrane complex
- Lytic cycle
- MAEBL, merozoite adhesive erythrocytic binding ligand
- MOI, multiplicity of infection
- OCRE, octamer repeat
- PDE, phosphodiesterase
- PKA, protein kinase A
- PKG, protein kinase G
- PM, plasma membrane
- QMEAN, Quality Model Energy Analysis
- Tachyzoite
- cAMP and cGMP signaling
- sgRNA, single guide RNA
- smHA, spaghetti monster-HA
Collapse
Affiliation(s)
- Kim Chi Vo
- Department of Molecular Parasitology, Institute of Biology, Faculty of Life Sciences, Humboldt University, Berlin, Germany
| | - Özlem Günay-Esiyok
- Department of Molecular Parasitology, Institute of Biology, Faculty of Life Sciences, Humboldt University, Berlin, Germany
| | - Nicolas Liem
- Experimental Biophysics, Institute of Biology, Faculty of Life Sciences, Humboldt University, Berlin, Germany
| | - Nishith Gupta
- Department of Molecular Parasitology, Institute of Biology, Faculty of Life Sciences, Humboldt University, Berlin, Germany.,Department of Biological Sciences, Birla Institute of Technology and Science Pilani (BITS-P), Hyderabad, India
| |
Collapse
|
49
|
Blake TCA, Haase S, Baum J. Actomyosin forces and the energetics of red blood cell invasion by the malaria parasite Plasmodium falciparum. PLoS Pathog 2020; 16:e1009007. [PMID: 33104759 PMCID: PMC7644091 DOI: 10.1371/journal.ppat.1009007] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/05/2020] [Accepted: 09/28/2020] [Indexed: 11/29/2022] Open
Abstract
All symptoms of malaria disease are associated with the asexual blood stages of development, involving cycles of red blood cell (RBC) invasion and egress by the Plasmodium spp. merozoite. Merozoite invasion is rapid and is actively powered by a parasite actomyosin motor. The current accepted model for actomyosin force generation envisages arrays of parasite myosins, pushing against short actin filaments connected to the external milieu that drive the merozoite forwards into the RBC. In Plasmodium falciparum, the most virulent human malaria species, Myosin A (PfMyoA) is critical for parasite replication. However, the precise function of PfMyoA in invasion, its regulation, the role of other myosins and overall energetics of invasion remain unclear. Here, we developed a conditional mutagenesis strategy combined with live video microscopy to probe PfMyoA function and that of the auxiliary motor PfMyoB in invasion. By imaging conditional mutants with increasing defects in force production, based on disruption to a key PfMyoA phospho-regulation site, the absence of the PfMyoA essential light chain, or complete motor absence, we define three distinct stages of incomplete RBC invasion. These three defects reveal three energetic barriers to successful entry: RBC deformation (pre-entry), mid-invasion initiation, and completion of internalisation, each requiring an active parasite motor. In defining distinct energetic barriers to invasion, these data illuminate the mechanical challenges faced in this remarkable process of protozoan parasitism, highlighting distinct myosin functions and identifying potential targets for preventing malaria pathogenesis.
Collapse
Affiliation(s)
- Thomas C. A. Blake
- Department of Life Sciences, Imperial College London, South Kensington, London, United Kingdom
| | - Silvia Haase
- Department of Life Sciences, Imperial College London, South Kensington, London, United Kingdom
| | - Jake Baum
- Department of Life Sciences, Imperial College London, South Kensington, London, United Kingdom
| |
Collapse
|
50
|
Anam ZE, Joshi N, Gupta S, Yadav P, Chaurasiya A, Kahlon AK, Kaushik S, Munde M, Ranganathan A, Singh S. A De novo Peptide from a High Throughput Peptide Library Blocks Myosin A -MTIP Complex Formation in Plasmodium falciparum. Int J Mol Sci 2020; 21:ijms21176158. [PMID: 32859024 PMCID: PMC7503848 DOI: 10.3390/ijms21176158] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/09/2020] [Accepted: 05/15/2020] [Indexed: 01/09/2023] Open
Abstract
Apicomplexan parasites, through their motor machinery, produce the required propulsive force critical for host cell-entry. The conserved components of this so-called glideosome machinery are myosin A and myosin A Tail Interacting Protein (MTIP). MTIP tethers myosin A to the inner membrane complex of the parasite through 20 amino acid-long C-terminal end of myosin A that makes direct contacts with MTIP, allowing the invasion of Plasmodium falciparum in erythrocytes. Here, we discovered through screening a peptide library, a de-novo peptide ZA1 that binds the myosin A tail domain. We demonstrated that ZA1 bound strongly to myosin A tail and was able to disrupt the native myosin A tail MTIP complex both in vitro and in vivo. We then showed that a shortened peptide derived from ZA1, named ZA1S, was able to bind myosin A and block parasite invasion. Overall, our study identified a novel anti-malarial peptide that could be used in combination with other antimalarials for blocking the invasion of Plasmodium falciparum.
Collapse
Affiliation(s)
- Zill e Anam
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
| | - Nishant Joshi
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, Uttar Pradesh 201304, India;
| | - Sakshi Gupta
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India; (S.G.); (M.M.)
| | - Preeti Yadav
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
| | - Ayushi Chaurasiya
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
| | - Amandeep Kaur Kahlon
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
| | - Shikha Kaushik
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
| | - Manoj Munde
- School of Physical Sciences, Jawaharlal Nehru University, New Delhi 110067, India; (S.G.); (M.M.)
| | - Anand Ranganathan
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
- Correspondence: (A.R.); (S.S.)
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India; (Z.e.A.); (P.Y.); (A.C.); (A.K.K.); (S.K.)
- Correspondence: (A.R.); (S.S.)
| |
Collapse
|