1
|
Mottaghi M, Karami P, Hesari Z, Nemati S, Mohammad Rahimi H, Mirjalali H. Evaluation of anti-Toxoplasma effects of solid lipid nanoparticles carrying Cinnamon zeylanicum and Moringa oleifera oil extracts. BMC Complement Med Ther 2024; 24:375. [PMID: 39449016 PMCID: PMC11515455 DOI: 10.1186/s12906-024-04677-x] [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: 04/20/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND The fabrication of anti-Toxoplasma drugs with less side effects and desirable efficacy is one of the important research goals facing with toxoplasmosis. This study aimed to determine the anti-Toxoplasma effects of Cinnamon zeylanicum (CZ), Moringa oleifera (MO) oil encapsulated into solid lipid nanoparticles (SLNs). METHODS Vero cells were cultured with serial concentrations (1 mg/mL to 100 µg/mL) of CZ-SLNs and MO-SLNs in DMEM culture medium. The morphological, physical, and chemical features of nanoparticles were calculated. The cell viability assays and anti-T. gondii effects of CZ-SLNs and MO-SLNs were evaluated. The CC50 and IC50 indices of SLNs-enveloped extracts were calculated. RESULTS The particle sizes of MO-SLNs and CZ-SLNs were 411.5 and 365 nm, while PDI indices were 0.53 and 0.7, respectively. Transmission electron microscopy (TEM) showed that both MO-SLNs and CZ-SLNs were smoothed spherical nanoparticles with rounded edges. The cytotoxicity assay showed the CC50 value of MO-SLNs at concentrations of ˃10 mg/mL. In addition, 60% of T. gondii-infected Vero cells remained alive at the concentrations ≤ 1 mg/ml, while the MO-SLNs killed at least 90% of T. gondii tachyzoites with an IC50 > 1 µg/ml. The cytotoxicity of CZ-SLNs extract showed the CC50 at the concentration 0.1 mg/mL. More than 50% of Vero cells, infected with T. gondii tachyzoites, survived at a concentration less than 0.1 mg/mL (IC50 ˂ 0.1 mg/mL), while the CZ-SLNs killed at least 85% of T. gondii tachyzoites in all concentrations. CONCLUSION The current results represented that the use of SLNs as a nano-carrier for M. oleifera and C. zeylanicum could kill T. gondii tachyzoites with low cytotoxicity, suggesting the effectiveness of these nano-emulsions along with the chemical agents in the treatment of Toxoplasma.
Collapse
Affiliation(s)
- Mahsa Mottaghi
- Department of Biology, Faculty of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Parisa Karami
- Department of Biology, Faculty of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Zahra Hesari
- Department of Pharmaceutics, School of Pharmacy, Guilan University of Medical Sciences, Guilan, Iran
| | - Sara Nemati
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hanieh Mohammad Rahimi
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Mirjalali
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Shi Y, Li X, Xue Y, Hu D, Song X. Cell cycle-regulated transcription factor AP2XII-9 is a key activator for asexual division and apicoplast inheritance in Toxoplasma gondii tachyzoite. mBio 2024; 15:e0133624. [PMID: 39207100 PMCID: PMC11481911 DOI: 10.1128/mbio.01336-24] [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: 05/01/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Toxoplasma gondii is an intracellular parasitic protozoan that poses a significant risk to the fetus carried by a pregnant woman or to immunocompromised individuals. T. gondii tachyzoites duplicate rapidly in host cells during acute infection through endodyogeny. This highly regulated division process is accompanied by complex gene regulation networks. TgAP2XII-9 is a cell cycle-regulated transcription factor, but its specific role in the parasite cell cycle is not fully understood. In this study, we demonstrate that TgAP2XII-9 is identified as a nuclear transcription factor and is dominantly expressed during the S/M phase of the tachyzoite cell cycle. Cleavage Under Targets and Tagmentation (CUT&Tag) results indicate that TgAP2XII-9 targets key genes for the moving junction machinery (RON2, 4, and 8) and daughter cell inner membrane complex (IMC). TgAP2XII-9 deficiency resulted in a significant downregulation of rhoptry proteins and rhoptry neck proteins, leading to a severe defect in the invasion and egress efficiency of tachyzoites. Additionally, the loss of TgAP2XII-9 correlated with a substantial downregulation of multiple IMC and apicoplast proteins, leading to disorders of daughter bud formation and apicoplast inheritance and further contributing to the inability of cell division and intracellular proliferation. Our study reveals that TgAP2XII-9 acts as a critical S/M-phase regulator that orchestrates the endodyogeny and apicoplast division in T. gondii tachyzoites. This study contributes to a broader understanding of the complexity of the parasite's cell cycle and its key regulators. IMPORTANCE The intracellular apicoplast parasite Toxoplasma gondii poses a great threat to the public health. The acute infection of T. gondii tachyzoites relies on efficient invasion by forming a moving junction structure and also fast replication by highly regulated endodyogeny. This study shows that an ApiAP2 transcription factor, TgAP2XII-9, acts as an activator for the S/M-phase gene expression, including genes related to daughter buds and moving junction formation. Loss of TgAP2XII-9 results in significant growth defects and disorders in endodyogeny and apicoplast inheritance of the parasites. Our results provide valuable insights into the transcriptional regulation of the parasite cell cycle and invading machinery in T. gondii.
Collapse
Affiliation(s)
- Yuehong Shi
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Xuan Li
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yingying Xue
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Dandan Hu
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, China
| | - Xingju Song
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning, China
| |
Collapse
|
3
|
Waller RF, Carruthers VB. Adaptations and metabolic evolution of myzozoan protists across diverse lifestyles and environments. Microbiol Mol Biol Rev 2024:e0019722. [PMID: 39387588 DOI: 10.1128/mmbr.00197-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024] Open
Abstract
SUMMARYMyzozoans encompass apicomplexans and dinoflagellates that manifest diverse lifestyles in highly varied environments. They show enormous propensity to employ different metabolic programs and exploit different nutrient resources and niches, and yet, they share much core biology that underlies this evolutionary success and impact. This review discusses apicomplexan parasites of medical significance and the traits and properties they share with non-pathogenic myzozoans. These include the versatility of myzozoan plastids, which scale from fully photosynthetic organelles to the site of very select key metabolic pathways. Pivotal evolutionary innovations, such as the apical complex, have allowed myzozoans to shift from predatory to parasitic and other symbiotic lifestyles multiple times in both apicomplexan and dinoflagellate branches of the myzozoan evolutionary tree. Such traits, along with shared mechanisms for nutrient acquisition, appear to underpin the prosperity of myzozoans in their varied habitats. Understanding the mechanisms of these shared traits has the potential to spawn new strategic interventions against medically and veterinary relevant parasites within this grouping.
Collapse
Affiliation(s)
- Ross F Waller
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Vern B Carruthers
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
4
|
Cabral G, Ren B, Bisio H, Otey D, Soldati-Favre D, Brown KM. Orthologs of Plasmodium ICM1 are dispensable for Ca 2+ mobilization in Toxoplasma gondii. Microbiol Spectr 2024; 12:e0122924. [PMID: 39162502 PMCID: PMC11448412 DOI: 10.1128/spectrum.01229-24] [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: 05/17/2024] [Accepted: 07/03/2024] [Indexed: 08/21/2024] Open
Abstract
Apicomplexan parasites mobilize ionic calcium (Ca2+) from intracellular stores to promote microneme secretion and facilitate motile processes including gliding motility, invasion, and egress. Recently, a multipass transmembrane protein, ICM1, was found to be important for calcium mobilization in Plasmodium falciparum and P. berghei. Comparative genomics and phylogenetics have revealed putative ICM orthologs in Toxoplasma gondii and other apicomplexans. T. gondii possesses two ICM-like proteins, which we have named TgICM1-L (TGGT1_305470) and TgICM2-L (TGGT1_309910). TgICM1-L and TgICM2-L localized to undefined puncta within the parasite cytosol. TgICM1-L and TgICM2-L are individually dispensable in tachyzoites, suggesting a potential compensatory relationship between the two proteins may exist. Surprisingly, mutants lacking both TgICM1-L and TgICM2-L are fully viable, exhibiting no obvious defects in growth, microneme secretion, invasion, or egress. Furthermore, loss of TgICM1-L, TgICM2-L, or both does not impair the parasite's ability to mobilize Ca2+. These findings suggest that additional proteins may participate in Ca2+ mobilization or import in Apicomplexa, reducing the dependence on ICM-like proteins in T. gondii. Collectively, these results highlight similar yet distinct mechanisms of Ca2+ mobilization between T. gondii and Plasmodium.IMPORTANCECa2+ signaling plays a crucial role in governing apicomplexan motility; yet, the mechanisms underlying Ca2+ mobilization from intracellular stores in these parasites remain unclear. In Plasmodium, the necessity of ICM1 for Ca2+ mobilization raises the question of whether this mechanism is conserved in other apicomplexans. Investigation into the orthologs of Plasmodium ICM1 in T. gondii revealed a differing requirement for ICM proteins between the two parasites. This study suggests that T. gondii employs ICM-independent mechanisms to regulate Ca2+ homeostasis and mobilization. Proteins involved in Ca2+ signaling in apicomplexans represent promising targets for therapeutic development.
Collapse
Affiliation(s)
- Gabriel Cabral
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Bingjian Ren
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Hugo Bisio
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
- Aix-Marseille Université, Centre National de la Recherche Scientifique, Information Génomique & Structurale, Marseille, France
| | - Dawson Otey
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Kevin M. Brown
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
5
|
Oliveira Souza RO, Yang C, Arrizabalaga G. Myosin A and F-Actin play a critical role in mitochondrial dynamics and inheritance in Toxoplasma gondii. PLoS Pathog 2024; 20:e1012127. [PMID: 39374269 PMCID: PMC11486366 DOI: 10.1371/journal.ppat.1012127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 10/17/2024] [Accepted: 09/23/2024] [Indexed: 10/09/2024] Open
Abstract
The single mitochondrion of the obligate intracellular parasite Toxoplasma gondii is highly dynamic. Toxoplasma's mitochondrion changes morphology as the parasite moves from the intracellular to the extracellular environment and during division. Toxoplasma's mitochondrial dynamic is dependent on an outer mitochondrion membrane-associated protein LMF1 and its interaction with IMC10, a protein localized at the inner membrane complex (IMC). In the absence of either LMF1 or IMC10, parasites have defective mitochondrial morphology and inheritance defects. As little is known about mitochondrial inheritance in Toxoplasma, we have used the LMF1/IMC10 tethering complex as an entry point to dissect the machinery behind this process. Using a yeast two-hybrid screen, we previously identified Myosin A (MyoA) as a putative interactor of LMF1. Although MyoA is known to be located at the parasite's pellicle, we now show through ultrastructure expansion microscopy (U-ExM) that this protein accumulates around the mitochondrion in the late stages of parasite division. Parasites lacking MyoA show defective mitochondrial morphology and a delay in mitochondrion delivery to the daughter parasite buds during division, indicating that this protein is involved in organellar inheritance. Disruption of the parasite's actin network also affects mitochondrion morphology. We also show that parasite-extracted mitochondrion vesicles interact with actin filaments. Interestingly, mitochondrion vesicles extracted out of parasites lacking LMF1 pulled down less actin, showing that LMF1 might be important for mitochondrion and actin interaction. Accordingly, we are showing for the first time that actin and Myosin A are important for Toxoplasma mitochondrial morphology and inheritance.
Collapse
Affiliation(s)
- Rodolpho Ornitz Oliveira Souza
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Chunlin Yang
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Gustavo Arrizabalaga
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| |
Collapse
|
6
|
Kent RS, Ward GE. Motility-dependent processes in Toxoplasma gondii tachyzoites and bradyzoites: same same but different. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.28.615543. [PMID: 39386639 PMCID: PMC11463423 DOI: 10.1101/2024.09.28.615543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The tachyzoite stage of the apicomplexan parasite Toxoplasma gondii utilizes motility for multiple purposes during its lytic cycle, including host cell invasion, egress from infected cells, and migration to new uninfected host cells to repeat the process. Bradyzoite stage parasites, which establish a new infection in a naïve host, must also use motility to escape from the cysts that are ingested by the new host and then migrate to the gut wall, where they either invade cells of the intestinal epithelium or squeeze between these cells to infect the underlying connective tissue. We know very little about the motility of bradyzoites, which we analyze in detail here and compare to the well-characterized motility and motility-dependent processes of tachyzoites. Unexpectedly, bradyzoites were found to be as motile as tachyzoites in a 3D model extracellular matrix, and they showed increased invasion into and transmigration across certain cell types, consistent with their need to establish the infection in the gut. The motility of the two stages was inhibited to the same extent by cytochalasin D and KNX-002, compounds known to target the parasite's actomyosin-based motor. In contrast, other compounds that impact tachyzoite motility (tachyplegin and enhancer 5) have less of an effect on bradyzoites, and rapid bradyzoite egress from infected cells is not triggered by treatment with calcium ionophores, as it is with tachyzoites. The similarities and differences between these two life cycle stages highlight the need to characterize both tachyzoites and bradyzoites for a more complete understanding of the role of motility in the parasite life cycle and the effect that potential therapeutics targeting parasite motility will have on disease establishment and progression.
Collapse
Affiliation(s)
- Robyn S Kent
- Department of Microbiology and Molecular Genetics, University of Vermont Larner College of Medicine, Burlington, Vermont, USA 05405
- 1041 BMSB, Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73190
| | - Gary E Ward
- Department of Microbiology and Molecular Genetics, University of Vermont Larner College of Medicine, Burlington, Vermont, USA 05405
| |
Collapse
|
7
|
Anaguano D, Adewale-Fasoro O, Vick GW, Yanik S, Blauwkamp J, Fierro MA, Absalon S, Srinivasan P, Muralidharan V. Plasmodium RON11 triggers biogenesis of the merozoite rhoptry pair and is essential for erythrocyte invasion. PLoS Biol 2024; 22:e3002801. [PMID: 39292724 PMCID: PMC11441699 DOI: 10.1371/journal.pbio.3002801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 09/30/2024] [Accepted: 08/13/2024] [Indexed: 09/20/2024] Open
Abstract
Malaria is a global and deadly human disease caused by the apicomplexan parasites of the genus Plasmodium. Parasite proliferation within human red blood cells (RBCs) is associated with the clinical manifestations of the disease. This asexual expansion within human RBCs begins with the invasion of RBCs by P. falciparum, which is mediated by the secretion of effectors from 2 specialized club-shaped secretory organelles in merozoite-stage parasites known as rhoptries. We investigated the function of the Rhoptry Neck Protein 11 (RON11), which contains 7 transmembrane domains and calcium-binding EF-hand domains. We generated conditional mutants of the P. falciparum RON11. Knockdown of RON11 inhibits parasite growth by preventing merozoite invasion. The loss of RON11 did not lead to any defects in processing of rhoptry proteins but instead led to a decrease in the amount of rhoptry proteins. We utilized ultrastructure expansion microscopy (U-ExM) to determine the effect of RON11 knockdown on rhoptry biogenesis. Surprisingly, in the absence of RON11, fully developed merozoites had only 1 rhoptry each. The single rhoptry in RON11-deficient merozoites were morphologically typical with a bulb and a neck oriented into the apical polar ring. Moreover, rhoptry proteins are trafficked accurately to the single rhoptry in RON11-deficient parasites. These data show that in the absence of RON11, the first rhoptry is generated during schizogony but upon the start of cytokinesis, the second rhoptry never forms. Interestingly, these single-rhoptry merozoites were able to attach to host RBCs but are unable to invade RBCs. Instead, RON11-deficient merozoites continue to engage with RBC for prolonged periods eventually resulting in echinocytosis, a result of secreting the contents from the single rhoptry into the RBC. Together, our data show that RON11 triggers the de novo biogenesis of the second rhoptry and functions in RBC invasion.
Collapse
Affiliation(s)
- David Anaguano
- Department of Cellular Biology, University of Georgia, Athens, Georgia, United States of America
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
| | - Opeoluwa Adewale-Fasoro
- Department of Molecular Microbiology and Immunology, and Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- The Johns Hopkins Malaria Research Institute, Baltimore, Maryland, United States of America
| | - Grace W. Vick
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Sean Yanik
- Department of Molecular Microbiology and Immunology, and Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- The Johns Hopkins Malaria Research Institute, Baltimore, Maryland, United States of America
| | - James Blauwkamp
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Manuel A. Fierro
- Department of Cellular Biology, University of Georgia, Athens, Georgia, United States of America
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
| | - Sabrina Absalon
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Prakash Srinivasan
- Department of Molecular Microbiology and Immunology, and Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- The Johns Hopkins Malaria Research Institute, Baltimore, Maryland, United States of America
| | - Vasant Muralidharan
- Department of Cellular Biology, University of Georgia, Athens, Georgia, United States of America
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
| |
Collapse
|
8
|
Back PS, Senthilkumar V, Choi CP, Quan JJ, Lou Q, Snyder AK, Ly AM, Lau JG, Zhou ZH, Ward GE, Bradley PJ. Alveolin proteins in the Toxoplasma inner membrane complex form a highly interconnected structure that maintains parasite shape and replication. PLoS Biol 2024; 22:e3002809. [PMID: 39264987 PMCID: PMC11421793 DOI: 10.1371/journal.pbio.3002809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 09/24/2024] [Accepted: 08/16/2024] [Indexed: 09/14/2024] Open
Abstract
Apicomplexan parasites possess several specialized structures to invade their host cells and replicate successfully. One of these is the inner membrane complex (IMC), a peripheral membrane-cytoskeletal system underneath the plasma membrane. It is composed of a series of flattened, membrane-bound vesicles and a cytoskeletal subpellicular network (SPN) comprised of intermediate filament-like proteins called alveolins. While the alveolin proteins are conserved throughout the Apicomplexa and the broader Alveolata, their precise functions and interactions remain poorly understood. Here, we describe the function of one of these alveolin proteins in Toxoplasma, IMC6. Disruption of IMC6 resulted in striking morphological defects that led to aberrant invasion and replication but surprisingly minor effects on motility. Deletion analyses revealed that the alveolin domain alone is largely sufficient to restore localization and partially sufficient for function. As this highlights the importance of the IMC6 alveolin domain, we implemented unnatural amino acid photoreactive crosslinking to the alveolin domain and identified multiple binding interfaces between IMC6 and 2 other cytoskeletal IMC proteins-IMC3 and ILP1. This provides direct evidence of protein-protein interactions in the alveolin domain and supports the long-held hypothesis that the alveolin domain is responsible for filament formation. Collectively, our study features the conserved alveolin proteins as critical components that maintain the parasite's structural integrity and highlights the alveolin domain as a key mediator of SPN architecture.
Collapse
Affiliation(s)
- Peter S Back
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Vignesh Senthilkumar
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Charles P Choi
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Justin J Quan
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Qing Lou
- Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Anne K Snyder
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, Vermont, United States of America
| | - Andrew M Ly
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Justin G Lau
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Z Hong Zhou
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, United States of America
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Gary E Ward
- Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, Vermont, United States of America
| | - Peter J Bradley
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California, United States of America
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
9
|
Liu Z, Li S, Anantha P, Thanakornsombut T, Wu L, Chen J, Tsuchiya R, Tripathi AK, Chen Y, Barman I. Plasmodium sporozoite shows distinct motility patterns in responses to three-dimensional environments. iScience 2024; 27:110463. [PMID: 39129829 PMCID: PMC11315120 DOI: 10.1016/j.isci.2024.110463] [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: 09/28/2023] [Revised: 11/20/2023] [Accepted: 07/02/2024] [Indexed: 08/13/2024] Open
Abstract
During malaria infection, Plasmodium sporozoites, the fast-moving stage of the parasite, are injected by a mosquito into the skin of the mammalian host. In the skin, sporozoites need to migrate through the dermal tissue to enter the blood vessel. Sporozoite motility is critical for infection but not well understood. Here, we used collagen hydrogels with tunable fiber structures, as an in vitro model for the skin. After injecting sporozoites into the hydrogel, we analyzed their motility in three-dimension (3D). We found that sporozoites demonstrated chiral motility, in that they mostly follow right-handed helical trajectories. In high-concentration collagen gel, sporozoites have lower instantaneous speed, but exhibit straighter tracks compared to low-concentration collagen gel, which leads to longer net displacement and faster dissemination. Taken together, our study indicates an inner mechanism for sporozoites to adapt to the environment, which could help with their successful exit from the skin tissue.
Collapse
Affiliation(s)
- Zhenhui Liu
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Songman Li
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Pooja Anantha
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | | | - Lintong Wu
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Junjie Chen
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Center for Cell Dynamics, Johns Hopkins University, Baltimore, MD, USA
| | - Ryohma Tsuchiya
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Center for Cell Dynamics, Johns Hopkins University, Baltimore, MD, USA
| | - Abhai K. Tripathi
- Department of Molecular Microbiology & Immunology, Johns Hopkins University, Baltimore, MD, USA
| | - Yun Chen
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Center for Cell Dynamics, Johns Hopkins University, Baltimore, MD, USA
| | - Ishan Barman
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
10
|
Yan Q, Gomis Perez C, Karatekin E. Cell Membrane Tension Gradients, Membrane Flows, and Cellular Processes. Physiology (Bethesda) 2024; 39:0. [PMID: 38501962 PMCID: PMC11368524 DOI: 10.1152/physiol.00007.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/18/2024] [Accepted: 03/18/2024] [Indexed: 03/20/2024] Open
Abstract
Cell membrane tension affects and is affected by many fundamental cellular processes, yet it is poorly understood. Recent experiments show that membrane tension can propagate at vastly different speeds in different cell types, reflecting physiological adaptations. Here we briefly review the current knowledge about membrane tension gradients, membrane flows, and their physiological context.
Collapse
Affiliation(s)
- Qi Yan
- Cellular and Molecular Physiology, Yale University, New Haven, Connecticut, United States
- Nanobiology Institute, Yale University, West Haven, Connecticut, United States
| | - Carolina Gomis Perez
- Cellular and Molecular Physiology, Yale University, New Haven, Connecticut, United States
- Nanobiology Institute, Yale University, West Haven, Connecticut, United States
| | - Erdem Karatekin
- Cellular and Molecular Physiology, Yale University, New Haven, Connecticut, United States
- Nanobiology Institute, Yale University, West Haven, Connecticut, United States
- Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, United States
- Wu Tsai Institute, Yale University, New Haven, Connecticut, United States
- Saints-Pères Paris Institute for the Neurosciences (SPPIN), Centre National de la Recherche Scientifique (CNRS), Paris, France
| |
Collapse
|
11
|
Brühlmann F, Perry C, Griessen C, Gunasekera K, Reymond JL, Naguleswaran A, Rottenberg S, Woods K, Olias P. TurboID mapping reveals the exportome of secreted intrinsically disordered proteins in the transforming parasite Theileria annulata. mBio 2024; 15:e0341223. [PMID: 38747635 PMCID: PMC11237503 DOI: 10.1128/mbio.03412-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/15/2024] [Indexed: 06/13/2024] Open
Abstract
Theileria annulata is a tick-transmitted apicomplexan parasite that gained the unique ability among parasitic eukaryotes to transform its host cell, inducing a fatal cancer-like disease in cattle. Understanding the mechanistic interplay between the host cell and malignant Theileria species that drives this transformation requires the identification of responsible parasite effector proteins. In this study, we used TurboID-based proximity labeling, which unbiasedly identified secreted parasite proteins within host cell compartments. By fusing TurboID to nuclear export or localization signals, we biotinylated proteins in the vicinity of the ligase enzyme in the nucleus or cytoplasm of infected macrophages, followed by mass spectrometry analysis. Our approach revealed with high confidence nine nuclear and four cytosolic candidate parasite proteins within the host cell compartments, eight of which had no orthologs in non-transforming T. orientalis. Strikingly, all eight of these proteins are predicted to be highly intrinsically disordered proteins. We discovered a novel tandem arrayed protein family, nuclear intrinsically disordered proteins (NIDP) 1-4, featuring diverse functions predicted by conserved protein domains. Particularly, NIDP2 exhibited a biphasic host cell-cycle-dependent localization, interacting with the EB1/CD2AP/CLASP1 parasite membrane complex at the schizont surface and the tumor suppressor stromal antigen 2 (STAG2), a cohesion complex subunit, in the host nucleus. In addition to STAG2, numerous NIDP2-associated host nuclear proteins implicated in various cancers were identified, shedding light on the potential role of the T. annulata exported protein family NIDP in host cell transformation and cancer-related pathways.IMPORTANCETurboID proximity labeling was used to identify secreted proteins of Theileria annulata, an apicomplexan parasite responsible for a fatal, proliferative disorder in cattle that represents a significant socio-economic burden in North Africa, central Asia, and India. Our investigation has provided important insights into the unique host-parasite interaction, revealing secreted parasite proteins characterized by intrinsically disordered protein structures. Remarkably, these proteins are conspicuously absent in non-transforming Theileria species, strongly suggesting their central role in the transformative processes within host cells. Our study identified a novel tandem arrayed protein family, with nuclear intrinsically disordered protein 2 emerging as a central player interacting with established tumor genes. Significantly, this work represents the first unbiased screening for exported proteins in Theileria and contributes essential insights into the molecular intricacies behind the malignant transformation of immune cells.
Collapse
Affiliation(s)
- Francis Brühlmann
- Institute of Animal Pathology, University of Bern, Bern, Switzerland
| | - Carmen Perry
- Institute of Animal Pathology, University of Bern, Bern, Switzerland
| | | | - Kapila Gunasekera
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, Bern, Switzerland
| | - Jean-Louis Reymond
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, Bern, Switzerland
| | | | - Sven Rottenberg
- Institute of Animal Pathology, University of Bern, Bern, Switzerland
| | - Kerry Woods
- Institute of Animal Pathology, University of Bern, Bern, Switzerland
| | - Philipp Olias
- Institute of Animal Pathology, University of Bern, Bern, Switzerland
- Institute of Veterinary Pathology, Justus Liebig University, Giessen, Germany
| |
Collapse
|
12
|
Katelas DA, Cruz-Miron R, Arroyo-Olarte RD, Brouwers JF, Srivastav RK, Gupta N. Phosphatidylserine synthase in the endoplasmic reticulum of Toxoplasma is essential for its lytic cycle in human cells. J Lipid Res 2024; 65:100535. [PMID: 38522751 PMCID: PMC11166882 DOI: 10.1016/j.jlr.2024.100535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 03/04/2024] [Accepted: 03/18/2024] [Indexed: 03/26/2024] Open
Abstract
Glycerophospholipids have emerged as a significant contributor to the intracellular growth of pathogenic protist Toxoplasma gondii. Phosphatidylserine (PtdSer) is one such lipid, attributed to the locomotion and motility-dependent invasion and egress events in its acutely infectious tachyzoite stage. However, the de novo synthesis of PtdSer and the importance of the pathway in tachyzoites remain poorly understood. We show that a base-exchange-type PtdSer synthase (PSS) located in the parasite's endoplasmic reticulum produces PtdSer, which is rapidly converted to phosphatidylethanolamine (PtdEtn) by PtdSer decarboxylase (PSD) activity. The PSS-PSD pathway enables the synthesis of several lipid species, including PtdSer (16:0/18:1) and PtdEtn (18:2/20:4, 18:1/18:2 and 18:2/22:5). The PSS-depleted strain exhibited a lower abundance of the major ester-linked PtdEtn species and concurrent accrual of host-derived ether-PtdEtn species. Most phosphatidylthreonine (PtdThr) species-an exclusive natural analog of PtdSer, also made in the endoplasmic reticulum-were repressed. PtdSer species, however, remained largely unaltered, likely due to the serine-exchange reaction of PtdThr synthase in favor of PtdSer upon PSS depletion. Not least, the loss of PSS abrogated the lytic cycle of tachyzoites, impairing the cell division, motility, and egress. In a nutshell, our data demonstrate a critical role of PSS in the biogenesis of PtdSer and PtdEtn species and its physiologically essential repurposing for the asexual reproduction of a clinically relevant intracellular pathogen.
Collapse
Affiliation(s)
- Dimitrios Alexandros Katelas
- Department of Molecular Parasitology, Faculty of Life Sciences, Humboldt University, Berlin, Germany; Intracellular Parasite Education and Research Labs (iPEARL), Department of Biological Sciences, Birla Institute of Technology and Science, Pilani (BITS-Pilani), Hyderabad, India
| | - Rosalba Cruz-Miron
- Department of Molecular Parasitology, Faculty of Life Sciences, Humboldt University, Berlin, Germany; Intracellular Parasite Education and Research Labs (iPEARL), Department of Biological Sciences, Birla Institute of Technology and Science, Pilani (BITS-Pilani), Hyderabad, India
| | - Ruben D Arroyo-Olarte
- Department of Molecular Parasitology, Faculty of Life Sciences, Humboldt University, Berlin, Germany; Carrera de Médico Cirujano y Unidad de Biomedicina (UBIMED), FES-Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Mexico
| | - Jos F Brouwers
- Analysis Techniques in the Life Sciences, Centre of Expertise Perspective in Health, Avans University of Applied Sciences, Breda, The Netherlands
| | - Ratnesh Kumar Srivastav
- Intracellular Parasite Education and Research Labs (iPEARL), Department of Biological Sciences, Birla Institute of Technology and Science, Pilani (BITS-Pilani), Hyderabad, India
| | - Nishith Gupta
- Department of Molecular Parasitology, Faculty of Life Sciences, Humboldt University, Berlin, Germany; Intracellular Parasite Education and Research Labs (iPEARL), Department of Biological Sciences, Birla Institute of Technology and Science, Pilani (BITS-Pilani), Hyderabad, India.
| |
Collapse
|
13
|
Ghosh A, Varshney A, Narwal SK, Nirdosh, Gupta R, Mishra S. The novel Plasmodium berghei protein S14 is essential for sporozoite gliding motility and infectivity. J Cell Sci 2024; 137:jcs261857. [PMID: 38832798 DOI: 10.1242/jcs.261857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 05/09/2024] [Indexed: 06/05/2024] Open
Abstract
Plasmodium sporozoites are the infective forms of the malaria parasite in the mosquito and vertebrate host. Gliding motility allows sporozoites to migrate and invade mosquito salivary glands and mammalian hosts. Motility and invasion are powered by an actin-myosin motor complex linked to the glideosome, which contains glideosome-associated proteins (GAPs), MyoA and the myosin A tail-interacting protein (MTIP). However, the role of several proteins involved in gliding motility remains unknown. We identified that the S14 gene is upregulated in sporozoite from transcriptome data of Plasmodium yoelii and further confirmed its transcription in P. berghei sporozoites using real-time PCR. C-terminal 3×HA-mCherry tagging revealed that S14 is expressed and localized on the inner membrane complex of the sporozoites. We disrupted S14 in P. berghei and demonstrated that it is essential for sporozoite gliding motility, and salivary gland and hepatocyte invasion. The gliding and invasion-deficient S14 knockout sporozoites showed normal expression and organization of inner membrane complex and surface proteins. Taken together, our data show that S14 plays a role in the function of the glideosome and is essential for malaria transmission.
Collapse
Affiliation(s)
- Ankit Ghosh
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Aastha Varshney
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Sunil Kumar Narwal
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Nirdosh
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Roshni Gupta
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Satish Mishra
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
14
|
Bento I, Parrington B, Pascual R, Goldberg AS, Wang E, Liu H, Zelle M, Takahashi JS, Elias JE, Mota MM, Rijo-Ferreira F. Circadian rhythms mediate malaria transmission potential. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594221. [PMID: 38798622 PMCID: PMC11118478 DOI: 10.1101/2024.05.14.594221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Malaria transmission begins when infected female Anopheles mosquitos deposit Plasmodium parasites into the mammalian host's skin during a bloodmeal. The salivary gland-resident sporozoite parasites migrate to the bloodstream, subsequently invading and replicating within hepatocytes. As Anopheles mosquitos are more active at night, with a 24-hour rhythm, we investigated whether their salivary glands are under circadian control, anticipating bloodmeals and modulating sporozoite biology for host encounters. Here we show that approximately half of the mosquito salivary gland transcriptome, particularly genes essential for efficient bloodmeals such as anti-blood clotting factors, exhibits circadian rhythmic expression. Furthermore, we demonstrate that mosquitoes prefer to feed during nighttime, with the amount of blood ingested varying cyclically throughout the day. Notably, we show a substantial subset of the sporozoite transcriptome cycling throughout the day. These include genes involved in parasite motility, potentially modulating the ability to initiate infection at different times of day. Thus, although sporozoites are typically considered quiescent, our results demonstrate their transcriptional activity, revealing robust daily rhythms of gene expression. Our findings suggest a circadian evolutionary relationship between the vector, parasite and mammalian host that together modulate malaria transmission.
Collapse
|
15
|
Currie-Olsen D, Leander BS. Novel cytoskeletal traits in the intestinal parasites (Squirmida, Platyproteum vivax) of Pacific peanut worms (Sipuncula, Phascolosoma agassizii). J Eukaryot Microbiol 2024; 71:e13023. [PMID: 38402546 DOI: 10.1111/jeu.13023] [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: 11/27/2023] [Revised: 01/18/2024] [Accepted: 02/11/2024] [Indexed: 02/26/2024]
Abstract
The cytoskeletal organization of a squirmid, namely Platyproteum vivax, was investigated with confocal laser scanning microscopy (CLSM) to refine inferences about convergent evolution among intestinal parasites of marine invertebrates. Platyproteum inhabits Pacific peanut worms (Phascolosoma agassizii) and has traits that are similar to other lineages of myzozoan parasites, namely gregarine apicomplexans within Selenidium, such as conspicuous feeding stages, called "trophozoites," capable of dynamic undulations. SEM and CLSM of P. vivax revealed an inconspicuous flagellar apparatus and a uniform array of longitudinal microtubules organized in bundles (LMBs). Extreme flattening of the trophozoites and a consistently oblique morphology of the anterior end provided a reliable way to distinguish dorsal and ventral surfaces. CLSM revealed a novel system of microtubules oriented in the flattened dorsoventral plane. Most of these dorsoventral microtubule bundles (DVMBs) had a punctate distribution and were evenly spaced along a curved line spanning the longitudinal axis of the trophozoites. This configuration of microtubules is inferred to function in maintaining the flattened shape of the trophozoites and facilitate dynamic undulations. The novel traits in Platyproteum are consistent with phylogenomic data showing that this lineage is only distantly related to Selenidium and other marine gregarine apicomplexans with dynamic intestinal trophozoites.
Collapse
Affiliation(s)
- Danja Currie-Olsen
- Department of Zoology, Beaty Biodiversity Research Centre and Museum, University of British Columbia, Vancouver, British Columbia, Canada
- Hakai Institute, Heriot Bay, Quadra Island, British Columbia, Canada
| | - Brian S Leander
- Department of Zoology, Beaty Biodiversity Research Centre and Museum, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
16
|
Chen R, Xu D, Zhao J, Tang X, Yang H, Liang H. Effects of cations on biofilms in gravity-driven membrane system: Filtration performance and mechanism investigation. WATER RESEARCH 2024; 254:121383. [PMID: 38432002 DOI: 10.1016/j.watres.2024.121383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/15/2024] [Accepted: 02/25/2024] [Indexed: 03/05/2024]
Abstract
The gravity-driven membrane (GDM) system is desirable for energy-efficient water treatment. However, little is known about the influence of cations on biofilm properties and GDM performance. In this study, typical cations (Ca2+ and Na+) were used to reveal the combined fouling behavior and mechanisms. Results showed that Ca2+ improved the stable flux and pollutant removal efficiency, while Na+ adversely affected the flux. Compared with GDM control, the concentration of pollutants was lower in Ca-GDM, as indicated by the low biomass, proteins, and polysaccharides. A heterogeneous and loose biofilm was observed in the Ca-GDM system, with roughness and porosity increasing by 43.06 % and 32.60 %, respectively. However, Na+ induced a homogeneous and dense biofilm, with porosity and roughness respectively reduced by 17.48 % and 22.04 %. The richness of bacterial communities increased in Ca-GDM systems, while it decreased in Na-GDM systems. High adenosine triphosphate (ATP) concentration in Ca-GDM system was consistent with the abundant bacteria and their high biological activity, which was helpful for the efficient removal of pollutants. The abundance of Apicomplexa, Platyhelminthes, Annelida and Nematoda increased after adding Ca2+, which was related to the formation of loose biofilms. Computational simulations indicated that the free volumes of the biofilms in Ca-GDM and Na-GDM were 13.7 and 13.2 nm3, respectively. The addition of cations changed intermolecular forces, Ca2+ induced bridging effects led to large and loose floc particles, while the significant dehydration of hydrated molecules in the Na-GDM caused obvious aggregation. Overall, microbiological characteristics and contaminant molecular interactions were the main reasons for differences in GDM systems.
Collapse
Affiliation(s)
- Rui Chen
- State Key Laboratory of Urban Water Resource and Environment (SKLUWRE), School of Environment, Harbin Institute of Technology, Harbin 150090, PR China; School of Environmental and Municipal Engineering, Xi'an University of Architecture and Technology, Xi'an 710055, PR China
| | - Daliang Xu
- State Key Laboratory of Urban Water Resource and Environment (SKLUWRE), School of Environment, Harbin Institute of Technology, Harbin 150090, PR China
| | - Jing Zhao
- State Key Laboratory of Urban Water Resource and Environment (SKLUWRE), School of Environment, Harbin Institute of Technology, Harbin 150090, PR China
| | - Xiaobin Tang
- State Key Laboratory of Urban Water Resource and Environment (SKLUWRE), School of Environment, Harbin Institute of Technology, Harbin 150090, PR China
| | - Haiyang Yang
- School of Civil Engineering, Guangzhou University, Guangzhou 510006, PR China
| | - Heng Liang
- State Key Laboratory of Urban Water Resource and Environment (SKLUWRE), School of Environment, Harbin Institute of Technology, Harbin 150090, PR China.
| |
Collapse
|
17
|
DePasquale JA. A comparison of teleost rodlet cells with apicomplexan cells. Acta Histochem 2024; 126:152167. [PMID: 38733697 DOI: 10.1016/j.acthis.2024.152167] [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: 01/17/2024] [Revised: 04/05/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024]
Abstract
Rodlet cells are unique pear-shaped cells found primarily in the epithelium of the teleost fishes. The rodlet cell was first identified by Thèlohan in 1892 who named it Rhabdospora thelohani as it was believed to be a protozoan parasite of the phylum Apicomplexa. The rodlet cell as parasite paradigm persisted for several decades afterwards but has since faded in the last 20 years or so. The rodlet cell is now generally believed to be an immune cell, functioning as an early responder to parasite intrusion. This short review makes a detailed comparison of apicomplexan structure and behavior with that of the rodlet cell to further strengthen the argument against a parasitic nature for the fish cell. It is then proposed that apical microvilli of the rodlet cell serve as a mechanical trigger for rodlet discharge as possible defense against larger ectoparasites.
Collapse
|
18
|
Qu Z, Li Y, Li W, Zhang N, Olajide JS, Mi X, Fu B. Global profiling of protein S-palmitoylation in the second-generation merozoites of Eimeria tenella. Parasitol Res 2024; 123:190. [PMID: 38647704 DOI: 10.1007/s00436-024-08204-2] [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: 11/18/2023] [Accepted: 04/04/2024] [Indexed: 04/25/2024]
Abstract
The intracellular protozoan Eimeria tenella is responsible for avian coccidiosis which is characterized by host intestinal damage. During developmental cycle, E. tenella undergoes versatile transitional stages such as oocyst, sporozoites, merozoites, and gametocytes. These developmental transitions involve changes in cell shape and cell size requiring cytoskeletal remodeling and changes in membrane proteins, which may require transcriptional and translational regulations as well as post-translational modification of proteins. Palmitoylation is a post-translational modification (PTM) of protein that orchestrates protein targeting, folding, stability, regulated enzymatic activity and even epigenetic regulation of gene expression. Previous research revealed that protein palmitoylation play essential role in Toxoplasma gondii, Trypanosoma cruzi, Trichomonas vaginalis, and several Plasmodium parasites. Until now, there is little information on the enzymes related to palmitoylation and role of protein acylation or palmitoylation in E. tenella. Therefore, palmitome of the second-generation merozoite of E. tenella was investigated. We identified a total of 2569 palmitoyl-sites that were assigned to 2145 palmitoyl-peptides belonging to 1561 protein-groups that participated in biological processes including parasite morphology, motility and host cell invasion. In addition, RNA biosynthesis, protein biosynthesis, folding, proteasome-ubiquitin degradation, and enzymes involved in PTMs, carbohydrate metabolism, glycan biosynthesis, and mitochondrial respiratory chain as well as vesicle trafficking were identified. The study allowed us to decipher the broad influence of palmitoylation in E. tenella biology, and its potential roles in the pathobiology of E. tenella infection. Raw data are publicly available at iProX with the dataset identifier PXD045061.
Collapse
Affiliation(s)
- Zigang Qu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
- Key Laboratory of Veterinary Public Health of the Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
- Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
- Jiangsu Co-Innovation Center for Prevention and Control of Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu Province, 225009, People's Republic of China
| | - Yuqiong Li
- Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan, 750002, People's Republic of China
| | - Wenhui Li
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
- Key Laboratory of Veterinary Public Health of the Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
- Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
- Jiangsu Co-Innovation Center for Prevention and Control of Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu Province, 225009, People's Republic of China
| | - Nianzhang Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
- Key Laboratory of Veterinary Public Health of the Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
- Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
- Jiangsu Co-Innovation Center for Prevention and Control of Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu Province, 225009, People's Republic of China
| | - Joshua Seun Olajide
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
- Key Laboratory of Veterinary Public Health of the Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
- Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China
| | - Xiaoyun Mi
- Xinjiang Key Laboratory of Animal Infectious Diseases, Institute of Veterinary Medicine, Xinjiang Academy of Animal Sciences, Urumqi, Xinjiang, 830013, People's Republic of China.
| | - Baoquan Fu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China.
- Key Laboratory of Veterinary Public Health of the Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China.
- Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province, 730046, People's Republic of China.
- Jiangsu Co-Innovation Center for Prevention and Control of Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu Province, 225009, People's Republic of China.
| |
Collapse
|
19
|
Ornitz Oliveira Souza R, Yang C, Arrizabalaga G. Myosin A and F-Actin play a critical role in mitochondrial dynamics and inheritance in Toxoplasma gondii. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.18.585462. [PMID: 38562694 PMCID: PMC10983951 DOI: 10.1101/2024.03.18.585462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The single mitochondrion of the obligate intracellular parasite Toxoplasma gondii is highly dynamic. Toxoplasma's mitochondrion changes morphology as the parasite moves from the intracellular to the extracellular environment and during division. Toxoplasma's mitochondrial dynamic is dependent on an outer mitochondrion membrane-associated protein LMF1 and its interaction with IMC10, a protein localized at the inner membrane complex (IMC). In the absence of either LMF1 or IMC10, parasites have defective mitochondrial morphology and inheritance defects. As little is known about mitochondrial inheritance in Toxoplasma, we have used the LMF1/IMC10 tethering complex as an entry point to dissect the machinery behind this process. Using a yeast two-hybrid screen, we previously identified Myosin A (MyoA) as a putative interactor of LMF1. Although MyoA is known to be located at the parasite's pellicle, we now show through ultrastructure expansion microscopy (U-ExM) that this protein accumulates around the mitochondrion in the late stages of parasite division. Parasites lacking MyoA show defective mitochondrial morphology and a delay in mitochondrion delivery to the daughter parasite buds during division, indicating that this protein is involved in organellar inheritance. Disruption of the parasite's actin network also affects mitochondrion morphology. We also show that parasite-extracted mitochondrion vesicles interact with actin filaments. Interestingly, mitochondrion vesicles extracted out of parasites lacking LMF1 pulled down less actin, showing that LMF1 might be important for mitochondrion and actin interaction. Accordingly, we are showing for the first time that actin and Myosin A are important for Toxoplasma mitochondrial morphology and inheritance.
Collapse
Affiliation(s)
| | - Chunlin Yang
- Department of Pharmacology and Toxicology, Indiana University School of Medicine
| | - Gustavo Arrizabalaga
- Department of Pharmacology and Toxicology, Indiana University School of Medicine
| |
Collapse
|
20
|
Suzuki-Tellier S, Kiørboe T, Simpson AGB. The function of the feeding groove of 'typical excavate' flagellates. J Eukaryot Microbiol 2024; 71:e13016. [PMID: 38108228 DOI: 10.1111/jeu.13016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/30/2023] [Accepted: 11/30/2023] [Indexed: 12/19/2023]
Abstract
Phagotrophic flagellates are the main consumers of bacteria and picophytoplankton. Despite their ecological significance in the 'microbial loop', many of their predation mechanisms remain unclear. 'Typical excavates' bear a ventral groove, where prey is captured for ingestion. The consequences of feeding through a 'semi-rigid' furrow on the prey size range have not been explored. An unidentified moving element called 'the wave' that sweeps along the bottom of the groove toward the site of phagocytosis has been observed in a few species; its function is unclear. We investigated the presence, behavior, and function of the wave in four species from the three excavate clades (Discoba, Metamonada, and Malawimonadida) and found it present in all studied cases, suggesting the potential homology of this feature across all three groups. The wave displayed a species-specific behavior and was crucial for phagocytosis. The morphology of the feeding groove had an upper-prey size limit for successful prey captures, but smaller particles were not constrained. Additionally, the ingestion efficiencies were species dependent. By jointly studying these feeding traits, we speculate on adaptations to differences in food availability to better understand their ecological functions.
Collapse
Affiliation(s)
- Sei Suzuki-Tellier
- Centre for Ocean Life, DTU Aqua, Technical University of Denmark, Kgs Lyngby, Denmark
| | - Thomas Kiørboe
- Centre for Ocean Life, DTU Aqua, Technical University of Denmark, Kgs Lyngby, Denmark
| | - Alastair G B Simpson
- Department of Biology, and Centre for Comparative Genomics and Evolutionary Bioinformatics, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
21
|
Yu Y, Zhang S, Duan C, Crouch C, Suo J, Tang X, Liu X, Liu J, Bruton B, Tarpey I, Suo X. Developing efficient strategies for localizing the enhanced yellow fluorescent protein subcellularly in transgenic Eimeria parasites. Sci Rep 2024; 14:4851. [PMID: 38418588 PMCID: PMC10902363 DOI: 10.1038/s41598-024-55569-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 02/25/2024] [Indexed: 03/01/2024] Open
Abstract
Eimeria species serve as promising eukaryotic vaccine vectors. And that the location of heterologous antigens in the subcellular components of genetically modified Eimeria may determine the magnitude and type of immune responses. Therefore, our study aimed to target a heterologous fluorescent protein to the cell surface or microneme, two locations where are more effective in inducing protective immunity, of Eimeria tenella and E. acervulina sporozoites. We used an enhanced yellow fluorescent protein (EYFP) as a tagging biomarker, fusing variously with some localization or whole sequences of compartmental proteins for targeting. After acquiring stable transgenic Eimeria populations, we observed EYFP expressing in expected locations with certain strategies. That is, EYFP successfully localized to the surface when it was fused between signal peptides and mature products of surface antigen 1 (SAG1). Furthermore, EYFP was efficiently targeted to the apical end, an optimal location for secretory organelle known as the microneme, when fused to the C terminus of microneme protein 2. Unexpectedly, EYFP exhibited dominantly in the apical end with only weak expression on the surface of the transgenic sporozoites when the parasites were transfected with plasmid with EYFP fused between signal peptides and mature products of E. tenella SAG 13. These strategies worked in both E. tenella and E. acervulina, laying a solid foundation for studying E. tenella and E. acervulina-based live vaccines that can be further tailored to the inclusion of cargo immunogens from other pathogens.
Collapse
Affiliation(s)
- Ying Yu
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, National Animal Protozoa Laboratory & College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Sixin Zhang
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, National Animal Protozoa Laboratory & College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Chunhui Duan
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, National Animal Protozoa Laboratory & College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Colin Crouch
- MSD Animal Health, Walton Manor, Milton Keynes, MK7 7AJ, UK
| | - Jingxia Suo
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, National Animal Protozoa Laboratory & College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Xinming Tang
- Key Laboratory of Animal Biosafety Risk Prevention and Control (North) of MARA, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xianyong Liu
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, National Animal Protozoa Laboratory & College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Jie Liu
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, National Animal Protozoa Laboratory & College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Beth Bruton
- MSD Animal Health, Walton Manor, Milton Keynes, MK7 7AJ, UK
| | - Ian Tarpey
- MSD Animal Health, Walton Manor, Milton Keynes, MK7 7AJ, UK
| | - Xun Suo
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, National Animal Protozoa Laboratory & College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
22
|
Anaguano D, Adewale-Fasoro O, Vick GS, Yanik S, Blauwkamp J, Fierro MA, Absalon S, Srinivasan P, Muralidharan V. Plasmodium RON11 triggers biogenesis of the merozoite rhoptry pair and is essential for erythrocyte invasion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.29.577654. [PMID: 38352500 PMCID: PMC10862748 DOI: 10.1101/2024.01.29.577654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Malaria is a global and deadly human disease caused by the apicomplexan parasites of the genus Plasmodium. Parasite proliferation within human red blood cells (RBC) is associated with the clinical manifestations of the disease. This asexual expansion within human RBCs, begins with the invasion of RBCs by P. falciparum, which is mediated by the secretion of effectors from two specialized club-shaped secretory organelles in merozoite-stage parasites known as rhoptries. We investigated the function of the Rhoptry Neck Protein 11 (RON11), which contains seven transmembrane domains and calcium-binding EF-hand domains. We generated conditional mutants of the P. falciparum RON11. Knockdown of RON11 inhibits parasite growth by preventing merozoite invasion. The loss of RON11 did not lead to any defects in processing of rhoptry proteins but instead led to a decrease in the amount of rhoptry proteins. We utilized ultrastructure expansion microscopy (U-ExM) to determine the effect of RON11 knockdown on rhoptry biogenesis. Surprisingly, in the absence of RON11, fully developed merozoites had only one rhoptry each. The single rhoptry in RON11 deficient merozoites were morphologically typical with a bulb and a neck oriented into the apical polar ring. Moreover, rhoptry proteins are trafficked accurately to the single rhoptry in RON11 deficient parasites. These data show that in the absence of RON11, the first rhoptry is generated during schizogony but upon the start of cytokinesis, the second rhoptry never forms. Interestingly, these single-rhoptry merozoites were able to attach to host RBCs but are unable to invade RBCs. Instead, RON11 deficient merozoites continue to engage with RBC for prolonged periods eventually resulting in echinocytosis, a result of secreting the contents from the single rhoptry into the RBC. Together, our data show that RON11 triggers the de novo biogenesis of the second rhoptry and functions in RBC invasion.
Collapse
Affiliation(s)
- David Anaguano
- Department of Cellular Biology, University of Georgia, Athens, GA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA
| | - Opeoluwa Adewale-Fasoro
- Department of Molecular Microbiology and Immunology, and Johns Hopkins Malaria Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Grace S. Vick
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA
| | - Sean Yanik
- Department of Molecular Microbiology and Immunology, and Johns Hopkins Malaria Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - James Blauwkamp
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis IN
| | - Manuel A. Fierro
- Department of Cellular Biology, University of Georgia, Athens, GA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA
| | - Sabrina Absalon
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis IN
| | - Prakash Srinivasan
- Department of Molecular Microbiology and Immunology, and Johns Hopkins Malaria Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Vasant Muralidharan
- Department of Cellular Biology, University of Georgia, Athens, GA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA
| |
Collapse
|
23
|
Yeh YT, Del Álamo JC, Caffrey CR. Biomechanics of parasite migration within hosts. Trends Parasitol 2024; 40:164-175. [PMID: 38172015 DOI: 10.1016/j.pt.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024]
Abstract
The dissemination of protozoan and metazoan parasites through host tissues is hindered by cellular barriers, dense extracellular matrices, and fluid forces in the bloodstream. To overcome these diverse biophysical impediments, parasites implement versatile migratory strategies. Parasite-exerted mechanical forces and upregulation of the host's cellular contractile machinery are the motors for these strategies, and these are comparably better characterized for protozoa than for helminths. Using the examples of the protozoans, Toxoplasma gondii and Plasmodium, and the metazoan, Schistosoma mansoni, we highlight how quantitative tools such as traction force and reflection interference contrast microscopies have improved our understanding of how parasites alter host mechanobiology to promote their migration.
Collapse
Affiliation(s)
- Yi-Ting Yeh
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98109, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA.
| | - Juan C Del Álamo
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98109, USA; Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA; Division of Cardiology, University of Washington, Seattle, WA 98109, USA; Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA 93093, USA
| | - Conor R Caffrey
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, 9500 Gilman Drive, MC0657, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
24
|
Rinkenberger N, Rosenberg A, Radke JB, Bhushan J, Tomita T, Weiss LM, Sibley LD. Susceptibility of Toxoplasma gondii to autophagy in human cells relies on multiple interacting parasite loci. mBio 2024; 15:e0259523. [PMID: 38095418 PMCID: PMC10790690 DOI: 10.1128/mbio.02595-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/06/2023] [Indexed: 01/04/2024] Open
Abstract
IMPORTANCE Autophagy is a process used by cells to recycle organelles and macromolecules and to eliminate intracellular pathogens. Previous studies have shown that some stains of Toxoplasma gondii are resistant to autophagy-dependent growth restriction, while others are highly susceptible. Although it is known that autophagy-mediated control requires activation by interferon gamma, the basis for why parasite strains differ in their susceptibility is unknown. Our findings indicate that susceptibility involves at least five unlinked parasite genes on different chromosomes, including several secretory proteins targeted to the parasite-containing vacuole and exposed to the host cell cytosol. Our findings reveal that susceptibility to autophagy-mediated growth restriction relies on differential recognition of parasite proteins exposed at the host-pathogen interface, thus identifying a new mechanism for cell-autonomous control of intracellular pathogens.
Collapse
Affiliation(s)
- Nicholas Rinkenberger
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Alex Rosenberg
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Joshua B. Radke
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Jaya Bhushan
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Tadakimi Tomita
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Louis M. Weiss
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - L. David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
25
|
Deng B, Vanagas L, Alonso AM, Angel SO. Proteomics Applications in Toxoplasma gondii: Unveiling the Host-Parasite Interactions and Therapeutic Target Discovery. Pathogens 2023; 13:33. [PMID: 38251340 PMCID: PMC10821451 DOI: 10.3390/pathogens13010033] [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: 11/13/2023] [Revised: 12/25/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
Toxoplasma gondii, a protozoan parasite with the ability to infect various warm-blooded vertebrates, including humans, is the causative agent of toxoplasmosis. This infection poses significant risks, leading to severe complications in immunocompromised individuals and potentially affecting the fetus through congenital transmission. A comprehensive understanding of the intricate molecular interactions between T. gondii and its host is pivotal for the development of effective therapeutic strategies. This review emphasizes the crucial role of proteomics in T. gondii research, with a specific focus on host-parasite interactions, post-translational modifications (PTMs), PTM crosstalk, and ongoing efforts in drug discovery. Additionally, we provide an overview of recent advancements in proteomics techniques, encompassing interactome sample preparation methods such as BioID (BirA*-mediated proximity-dependent biotin identification), APEX (ascorbate peroxidase-mediated proximity labeling), and Y2H (yeast two hybrid), as well as various proteomics approaches, including single-cell analysis, DIA (data-independent acquisition), targeted, top-down, and plasma proteomics. Furthermore, we discuss bioinformatics and the integration of proteomics with other omics technologies, highlighting its potential in unraveling the intricate mechanisms of T. gondii pathogenesis and identifying novel therapeutic targets.
Collapse
Affiliation(s)
- Bin Deng
- Department of Biology and VBRN Proteomics Facility, University of Vermont, Burlington, VT 05405, USA
| | - Laura Vanagas
- Laboratorio de Parasitología Molecular, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Chascomús 7130, Provincia de Buenos Aires, Argentina; (L.V.); (S.O.A.); (A.M.A.)
- Escuela de Bio y Nanotecnologías (UNSAM), 25 de Mayo y Francia. C.P., San Martín 1650, Provincia de Buenos Aires, Argentina
| | - Andres M. Alonso
- Laboratorio de Parasitología Molecular, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Chascomús 7130, Provincia de Buenos Aires, Argentina; (L.V.); (S.O.A.); (A.M.A.)
- Escuela de Bio y Nanotecnologías (UNSAM), 25 de Mayo y Francia. C.P., San Martín 1650, Provincia de Buenos Aires, Argentina
| | - Sergio O. Angel
- Laboratorio de Parasitología Molecular, Instituto Tecnológico de Chascomús (CONICET-UNSAM), Chascomús 7130, Provincia de Buenos Aires, Argentina; (L.V.); (S.O.A.); (A.M.A.)
- Escuela de Bio y Nanotecnologías (UNSAM), 25 de Mayo y Francia. C.P., San Martín 1650, Provincia de Buenos Aires, Argentina
| |
Collapse
|
26
|
Bondoc-Naumovitz KG, Laeverenz-Schlogelhofer H, Poon RN, Boggon AK, Bentley SA, Cortese D, Wan KY. Methods and Measures for Investigating Microscale Motility. Integr Comp Biol 2023; 63:1485-1508. [PMID: 37336589 PMCID: PMC10755196 DOI: 10.1093/icb/icad075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/31/2023] [Accepted: 06/06/2023] [Indexed: 06/21/2023] Open
Abstract
Motility is an essential factor for an organism's survival and diversification. With the advent of novel single-cell technologies, analytical frameworks, and theoretical methods, we can begin to probe the complex lives of microscopic motile organisms and answer the intertwining biological and physical questions of how these diverse lifeforms navigate their surroundings. Herein, we summarize the main mechanisms of microscale motility and give an overview of different experimental, analytical, and mathematical methods used to study them across different scales encompassing the molecular-, individual-, to population-level. We identify transferable techniques, pressing challenges, and future directions in the field. This review can serve as a starting point for researchers who are interested in exploring and quantifying the movements of organisms in the microscale world.
Collapse
Affiliation(s)
| | | | - Rebecca N Poon
- Living Systems Institute, University of Exeter, Stocker Road, EX4 4QD, Exeter, UK
| | - Alexander K Boggon
- Living Systems Institute, University of Exeter, Stocker Road, EX4 4QD, Exeter, UK
| | - Samuel A Bentley
- Living Systems Institute, University of Exeter, Stocker Road, EX4 4QD, Exeter, UK
| | - Dario Cortese
- Living Systems Institute, University of Exeter, Stocker Road, EX4 4QD, Exeter, UK
| | - Kirsty Y Wan
- Living Systems Institute, University of Exeter, Stocker Road, EX4 4QD, Exeter, UK
| |
Collapse
|
27
|
Rodriguez JB, Szajnman SH. An updated review of chemical compounds with anti-Toxoplasma gondii activity. Eur J Med Chem 2023; 262:115885. [PMID: 37871407 DOI: 10.1016/j.ejmech.2023.115885] [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: 08/24/2023] [Revised: 09/30/2023] [Accepted: 10/15/2023] [Indexed: 10/25/2023]
Abstract
The opportunistic apicomplexan parasite Toxoplasma gondii is the etiologic agent for toxoplasmosis, which can infect a widespread range of hosts, particularly humans and warm-blooded animals. The present chemotherapy to treat or prevent toxoplasmosis is deficient and is based on diverse drugs such as atovaquone, trimethoprim, spiramycine, which are effective in acute toxoplasmosis. Therefore, a safe chemotherapy is required for toxoplasmosis considering that its responsible agent, T. gondii, provokes severe illness and death in pregnant women and immunodeficient patients. A certain disadvantage of the available treatments is the lack of effectiveness against the tissue cyst of the parasite. A safe chemotherapy to combat toxoplasmosis should be based on the metabolic differences between the parasite and the mammalian host. This article covers different relevant molecular targets to combat this disease including the isoprenoid pathway (farnesyl diphosphate synthase, squalene synthase), dihydrofolate reductase, calcium-dependent protein kinases, histone deacetylase, mitochondrial electron transport chain, etc.
Collapse
Affiliation(s)
- Juan B Rodriguez
- Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, C1428EHA, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Unidad de Microanálisis y Métodos Físicos en Química Orgánica (UMYMFOR), C1428EHA, Buenos Aires, Argentina.
| | - Sergio H Szajnman
- Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pabellón 2, C1428EHA, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Unidad de Microanálisis y Métodos Físicos en Química Orgánica (UMYMFOR), C1428EHA, Buenos Aires, Argentina
| |
Collapse
|
28
|
Zhang Z, Gaetjens TK, Ou J, Zhou Q, Yu Y, Mallory DP, Abel SM, Yu Y. Propulsive cell entry diverts pathogens from immune degradation by remodeling the phagocytic synapse. Proc Natl Acad Sci U S A 2023; 120:e2306788120. [PMID: 38032935 PMCID: PMC10710034 DOI: 10.1073/pnas.2306788120] [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: 05/01/2023] [Accepted: 10/05/2023] [Indexed: 12/02/2023] Open
Abstract
Phagocytosis is a critical immune function for infection control and tissue homeostasis. During phagocytosis, pathogens are internalized and degraded in phagolysosomes. For pathogens that evade immune degradation, the prevailing view is that virulence factors are required to disrupt the biogenesis of phagolysosomes. In contrast, we present here that physical forces from motile pathogens during cell entry divert them away from the canonical degradative pathway. This altered fate begins with the force-induced remodeling of the phagocytic synapse formation. We used the parasite Toxoplasma gondii as a model because live Toxoplasma actively invades host cells using gliding motility. To differentiate the effects of physical forces from virulence factors in phagocytosis, we employed magnetic forces to induce propulsive entry of inactivated Toxoplasma into macrophages. Experiments and computer simulations show that large propulsive forces hinder productive activation of receptors by preventing their spatial segregation from phosphatases at the phagocytic synapse. Consequently, the inactivated parasites are engulfed into vacuoles that fail to mature into degradative units, similar to the live motile parasite's intracellular pathway. Using yeast cells and opsonized beads, we confirmed that this mechanism is general, not specific to the parasite used. These results reveal new aspects of immune evasion by demonstrating how physical forces during active cell entry, independent of virulence factors, enable pathogens to circumvent phagolysosomal degradation.
Collapse
Affiliation(s)
- Zihan Zhang
- Department of Chemistry, Indiana University, Bloomington, IN47405-7102
| | - Thomas K. Gaetjens
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN37996
| | - Jin Ou
- Department of Chemistry, Indiana University, Bloomington, IN47405-7102
| | - Qiong Zhou
- Department of Chemistry, Indiana University, Bloomington, IN47405-7102
| | - Yanqi Yu
- Department of Chemistry, Indiana University, Bloomington, IN47405-7102
| | - D. Paul Mallory
- Department of Chemistry, Indiana University, Bloomington, IN47405-7102
| | - Steven M. Abel
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN37996
| | - Yan Yu
- Department of Chemistry, Indiana University, Bloomington, IN47405-7102
| |
Collapse
|
29
|
Yin D, Jiang N, Cheng C, Sang X, Feng Y, Chen R, Chen Q. Protein Lactylation and Metabolic Regulation of the Zoonotic Parasite Toxoplasma gondii. GENOMICS, PROTEOMICS & BIOINFORMATICS 2023; 21:1163-1181. [PMID: 36216028 PMCID: PMC11082259 DOI: 10.1016/j.gpb.2022.09.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 09/06/2022] [Accepted: 09/29/2022] [Indexed: 06/16/2023]
Abstract
The biology of Toxoplasma gondii, the causative pathogen of one of the most widespread parasitic diseases (toxoplasmosis), remains poorly understood. Lactate, which is derived from glucose metabolism, is not only an energy source in a variety of organisms, including T. gondii, but also a regulatory molecule that participates in gene activation and protein function. Lysine lactylation (Kla) is a type of post-translational modifications (PTMs) that has been recently associated with chromatin remodeling; however, Kla of histone and non-histone proteins has not yet been studied in T. gondii. To examine the prevalence and function of lactylation in T. gondii parasites, we mapped the lactylome of proliferating tachyzoite cells and identified 1964 Kla sites on 955 proteins in the T. gondii RH strain. Lactylated proteins were distributed in multiple subcellular compartments and were closely related to a wide variety of biological processes, including mRNA splicing, glycolysis, aminoacyl-tRNA biosynthesis, RNA transport, and many signaling pathways. We also performed a chromatin immunoprecipitation sequencing (ChIP-seq) analysis using a lactylation-specific antibody and found that the histones H4K12la and H3K14la were enriched in the promoter and exon regions of T. gondii associated with microtubule-based movement and cell invasion. We further confirmed the delactylase activity of histone deacetylases TgHDAC2-4, and found that treatment with anti-histone acetyltransferase (TgMYST-A) antibodies profoundly reduced protein lactylation in T. gondii. This study offers the first dataset of the global lactylation proteome and provides a basis for further dissecting the functional biology of T. gondii.
Collapse
Affiliation(s)
- Deqi Yin
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110166, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang 110866, China
| | - Ning Jiang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110166, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang 110866, China
| | - Chang Cheng
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110166, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang 110866, China
| | - Xiaoyu Sang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110166, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang 110866, China
| | - Ying Feng
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110166, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang 110866, China
| | - Ran Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110166, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang 110866, China
| | - Qijun Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110166, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang 110866, China.
| |
Collapse
|
30
|
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
|
31
|
Valigurová A, Diakin A, Seifertová M, Vaškovicová N, Kováčiková M, Paskerova GG. Dispersal and invasive stages of Urospora eugregarines (Apicomplexa) from brown bodies of a polychaete host. J Invertebr Pathol 2023; 201:107997. [PMID: 37774965 DOI: 10.1016/j.jip.2023.107997] [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: 07/18/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023]
Abstract
Urosporid eugregarines (Apicomplexa: Urosporidae) are unicellular eukaryotic parasites inhabiting the coelom or the intestine of marine invertebrates such as annelids, molluscs, nemerteans, and echinoderms. Despite the availability of published morphological and phylogenetical analyses of coelomic gregarines, their long-term survival in the host body cavity and dispersal routes into the marine environment remain unclear. Here, we focus on Urospora gametocysts and oocysts with sporozoites, which were found viable inside the so-called brown bodies floating in the body cavity of the polychaete Travisia forbesii. Brown bodies form as a result of host defence where coelomocytes encapsulate dead host cells and foreign objects including potential pathogens. We hypothesise the long-term persistence of Urospora eugregarines in brown bodies through evasion of the host immunity and outline possible pathways for their egress into the marine environment, applicable as dispersal routes for other parasites as well. Unique features revealed by detailed ultrastructural analysis of detected eugregarine stages include asynchronous sporogony, a massive sporozoite secretion apparatus, as well as the presence of free (possibly autoinfective) sporozoites within the gametocyst. The assignment to the genus Urospora and the complete identity with U. ovalis and U. travisiae were confirmed by analysing 18S rDNA sequences obtained from isolated gametocysts. The 18S rDNA phylogeny confirmed the affiliation of Urosporidae to Lecudinoidea and the grouping of all Urospora sequences with Difficilina from nemerteans and environmental sequences from the Artic region. We also enriched the Apicomplexa set by partial 28S rDNA sequences of two Urospora species enabling more complex phylogenetic analyses prospectively.
Collapse
Affiliation(s)
- Andrea Valigurová
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic.
| | - Andrei Diakin
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic
| | - Mária Seifertová
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic
| | - Naděžda Vaškovicová
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Magdaléna Kováčiková
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic
| | - Gita G Paskerova
- Department of Invertebrate Zoology, Faculty of Biology, Saint Petersburg State University, Universitetskaya emb. 7/9, St Petersburg 199034, Russian Federation
| |
Collapse
|
32
|
Rawat RS, Gupta A, Antil N, Bhatnagar S, Singh M, Rawat A, Prasad TSK, Sharma P. Protein kinase PfPK2 mediated signalling is critical for host erythrocyte invasion by malaria parasite. PLoS Pathog 2023; 19:e1011770. [PMID: 37988347 PMCID: PMC10662742 DOI: 10.1371/journal.ppat.1011770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 10/23/2023] [Indexed: 11/23/2023] Open
Abstract
Signalling pathways in malaria parasite remain poorly defined and major reason for this is the lack of understanding of the function of majority of parasite protein kinases and phosphatases in parasite signalling and its biology. In the present study, we have elucidated the function of Protein Kinase 2 (PfPK2), which is known to be indispensable for the survival of human malaria parasite Plasmodium falciparum. We demonstrate that it is involved in the invasion of host erythrocytes, which is critical for establishing infection. In addition, PfPK2 may also be involved in the maturation of the parasite post-invasion. PfPK2 regulates the release of microneme proteins like Apical Membrane Antigen 1 (AMA1), which facilitates the formation of Tight Junction between the merozoite and host erythrocyte- a key step in the process of invasion. Comparative phosphoproteomics studies revealed that PfPK2 may be involved in regulation of several key proteins involved in invasion and signalling. Furthermore, PfPK2 regulates the generation of cGMP and the release of calcium in the parasite, which are key second messengers for the process of invasion. These and other studies have shed light on a novel signalling pathway in which PfPK2 acts as an upstream regulator of important cGMP-calcium signalling, which plays an important role in parasite invasion.
Collapse
Affiliation(s)
- Rahul Singh Rawat
- Eukaryotic Gene Expression Laboratory, National Institute of Immunology, New Delhi, India
| | - Ankit Gupta
- Eukaryotic Gene Expression Laboratory, National Institute of Immunology, New Delhi, India
| | - Neelam Antil
- Institute of Bioinformatics, International Tech Park, Bangalore, India
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, India
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Sonika Bhatnagar
- Eukaryotic Gene Expression Laboratory, National Institute of Immunology, New Delhi, India
| | - Monika Singh
- Eukaryotic Gene Expression Laboratory, National Institute of Immunology, New Delhi, India
| | - Akanksha Rawat
- Eukaryotic Gene Expression Laboratory, National Institute of Immunology, New Delhi, India
| | - T. S. Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Pushkar Sharma
- Eukaryotic Gene Expression Laboratory, National Institute of Immunology, New Delhi, India
| |
Collapse
|
33
|
Fritz-Laylin LK, Titus MA. The evolution and diversity of actin-dependent cell migration. Mol Biol Cell 2023; 34:pe6. [PMID: 37906436 PMCID: PMC10846614 DOI: 10.1091/mbc.e22-08-0358] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 11/02/2023] Open
Abstract
Many eukaryotic cells, including animal cells and unicellular amoebae, use dynamic-actin networks to crawl across solid surfaces. Recent discoveries of actin-dependent crawling in additional lineages have sparked interest in understanding how and when this type of motility evolved. Tracing the evolution of cell crawling requires understanding the molecular mechanisms underlying motility. Here we outline what is known about the diversity and evolution of the molecular mechanisms that drive cell motility, with a focus on actin-dependent crawling. Classic studies and recent work have revealed a surprising number of distinct mechanical modes of actin-dependent crawling used by different cell types and species to navigate different environments. The overlap in actin network regulators driving multiple types of actin-dependent crawling, along with cortical-actin networks that support the plasma membrane in these cells, suggest that actin motility and cortical actin networks might have a common evolutionary origin. The rapid development of additional evolutionarily diverse model systems, advanced imaging technologies, and CRISPR-based genetic tools, is opening the door to testing these and other new ideas about the evolution of actin-dependent cell crawling.
Collapse
Affiliation(s)
| | - Margaret A. Titus
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
34
|
Cruz-Bustos T, Dolezal M, Feix AS, Ruttkowski B, Hummel K, Razzazi-Fazeli E, Joachim A. Unravelling the sexual developmental biology of Cystoisospora suis, a model for comparative coccidian parasite studies. Front Cell Infect Microbiol 2023; 13:1271731. [PMID: 37953800 PMCID: PMC10635411 DOI: 10.3389/fcimb.2023.1271731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/28/2023] [Indexed: 11/14/2023] Open
Abstract
Introduction The apicomplexan parasite Cystoisospora suis has global significance as an enteropathogen of suckling piglets. Its intricate life cycle entails a transition from an asexual phase to sexual development, ultimately leading to the formation of transmissible oocysts. Methods To advance our understanding of the parasite's cellular development, we complemented previous transcriptome studies by delving into the proteome profiles at five distinct time points of in vitro cultivation through LC/MS-MS analysis. Results A total of 1,324 proteins were identified in the in vitro developmental stages of C. suis, and 1,082 proteins were identified as significantly differentially expressed. Data are available via ProteomeXchange with identifier PXD045050. We performed BLAST, GO enrichment, and KEGG pathway analyses on the up- and downregulated proteins to elucidate correlated events in the C. suis life cycle. Our analyses revealed intriguing metabolic patterns in macromolecule metabolism, DNA- and RNA-related processes, proteins associated with sexual stages, and those involved in cell invasion, reflecting the adaptation of sexual stages to a nutrient-poor and potentially stressful extracellular environment, with a focus on enzymes involved in metabolism and energy production. Discussion These findings have important implications for understanding the developmental biology of C. suis as well as other, related coccidian parasites, such as Eimeria spp. and Toxoplasma gondii. They also support the role of C. suis as a new model for the comparative biology of coccidian tissue cyst stages.
Collapse
Affiliation(s)
- Teresa Cruz-Bustos
- Department of Pathobiology, Institute of Parasitology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Marlies Dolezal
- Platform for Bioinformatics and Biostatistics, Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Anna Sophia Feix
- Department of Pathobiology, Institute of Parasitology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Bärbel Ruttkowski
- Department of Pathobiology, Institute of Parasitology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Karin Hummel
- VetCore Facility (Proteomics), University of Veterinary Medicine Vienna, Vienna, Austria
| | - Ebrahim Razzazi-Fazeli
- VetCore Facility (Proteomics), University of Veterinary Medicine Vienna, Vienna, Austria
| | - Anja Joachim
- Department of Pathobiology, Institute of Parasitology, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
35
|
Agrawal P, Kumari S, Mohmmed A, Malhotra P, Sharma U, Sahal D. Identification of Novel, Potent, and Selective Compounds against Malaria Using Glideosomal-Associated Protein 50 as a Drug Target. ACS OMEGA 2023; 8:38506-38523. [PMID: 37867646 PMCID: PMC10586260 DOI: 10.1021/acsomega.3c05323] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 09/01/2023] [Indexed: 10/24/2023]
Abstract
Phylum apicomplexan consists of parasites, such as Plasmodium and Toxoplasma. These obligate intracellular parasites enter host cells via an energy-dependent process using specialized machinery, called the glideosome. In the present study, we used Plasmodium falciparum GAP50, a glideosome-associated protein, as a target to screen 951 different compounds from diverse chemical libraries. Using different screening methods, eight compounds (Hayatinine, Curine, MMV689758 (Bedaquiline), MMV1634402 (Brilacidin), and MMV688271, MMV782353, MMV642550, and USINB4-124-8) were identified, which showed promising binding affinity (KD < 75 μM), along with submicromolar range antiparasitic efficacy and selectivity index > 100 fold for malaria parasite. These eight compounds were effective against Chloroquine-resistant PfINDO and Artemisinin-resistant PfCam3.1R359T strains. Studies on the effect of these compounds at asexual blood stages showed that these eight compounds act differently at different developmental stages, indicating the binding of these compounds to other Plasmodium proteins, in addition to PfGAP50. We further studied the effects of compounds (Bedaquiline and USINB4-124-8) in an in vivoPlasmodium berghei mouse model of malaria. Importantly, the oral delivery of Bedaquiline (50 mg/kg b. wt.) showed substantial suppression of parasitemia, and three out of seven mice were cured of the infection. Thus, our study provides new scaffolds for the development of antimalarials that can act at multiple Plasmodium lifecycle stages.
Collapse
Affiliation(s)
- Prakhar Agrawal
- International
Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Surekha Kumari
- Chemical
Technology Division, CSIR-Institute of Himalayan
Bioresource Technology, Palampur 176061, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Asif Mohmmed
- International
Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Pawan Malhotra
- International
Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Upendra Sharma
- Chemical
Technology Division, CSIR-Institute of Himalayan
Bioresource Technology, Palampur 176061, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Dinkar Sahal
- International
Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| |
Collapse
|
36
|
Boisard J, Florent I. Porospora gigantea. Trends Parasitol 2023; 39:886-887. [PMID: 37344291 DOI: 10.1016/j.pt.2023.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 06/23/2023]
Affiliation(s)
- Julie Boisard
- Département Adaptations du Vivant (AVIV), Molécules de Communication et Adaptation des Microorganismes (MCAM, UMR 7245 CNRS), Muséum National d'Histoire Naturelle, CNRS, CP 52, 57 rue Cuvier, 75231 Cedex 05 Paris, France
| | - Isabelle Florent
- Département Adaptations du Vivant (AVIV), Molécules de Communication et Adaptation des Microorganismes (MCAM, UMR 7245 CNRS), Muséum National d'Histoire Naturelle, CNRS, CP 52, 57 rue Cuvier, 75231 Cedex 05 Paris, France.
| |
Collapse
|
37
|
Wang QQ, Sun M, Tang T, Lai DH, Liu J, Maity S, He K, Wu XT, Yang J, Li YB, Tang XY, Ding HY, Hide G, Distefano M, Lun ZR, Zhu XQ, Long S. Functional screening reveals Toxoplasma prenylated proteins required for endocytic trafficking and rhoptry protein sorting. mBio 2023; 14:e0130923. [PMID: 37548452 PMCID: PMC10470541 DOI: 10.1128/mbio.01309-23] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/12/2023] [Indexed: 08/08/2023] Open
Abstract
In the apicomplexans, endocytosed cargos (e.g., hemoglobin) are trafficked to a specialized organelle for digestion. This follows a unique endocytotic process at the micropore/cytostome in these parasites. However, the mechanism underlying endocytic trafficking remains elusive, due to the repurposing of classical endocytic proteins for the biogenesis of apical organelles. To resolve this issue, we have exploited the genetic tractability of the model apicomplexan Toxoplasma gondii, which ingests host cytosolic materials (e.g., green fluorescent protein[GFP]). We determined an association between protein prenylation and endocytic trafficking, and using an alkyne-labeled click chemistry approach, the prenylated proteome was characterized. Genome editing, using clustered regularly interspaced short palindromic repaet/CRISPR-associated nuclease 9 (CRISPR/Cas9), was efficiently utilized to generate genetically modified lines for the functional screening of 23 prenylated candidates. This identified four of these proteins that regulate the trafficking of endocytosed GFP vesicles. Among these proteins, Rab1B and YKT6.1 are highly conserved but are non-classical endocytic proteins in eukaryotes. Confocal imaging analysis showed that Rab1B and Ras are substantially localized to both the trans-Golgi network and the endosome-like compartments in the parasite. Conditional knockdown of Rab1B caused a rapid defect in secretory trafficking to the rhoptry bulb, suggesting a trafficking intersection role for the key regulator Rab1B. Further experiments confirmed a critical role for protein prenylation in regulating the stability/activity of these proteins (i.e., Rab1B and YKT6.1) in the parasite. Our findings define the molecular basis of endocytic trafficking and reveal a potential intersection function of Rab1B on membrane trafficking in T. gondii. This might extend to other related protists, including the malarial parasites. IMPORTANCE The protozoan Toxoplasma gondii establishes a permissive niche, in host cells, that allows parasites to acquire large molecules such as proteins. Numerous studies have demonstrated that the parasite repurposes the classical endocytic components for secretory sorting to the apical organelles, leaving the question of endocytic transport to the lysosome-like compartment unclear. Recent studies indicated that endocytic trafficking is likely to associate with protein prenylation in malarial parasites. This information promoted us to examine this association in the model apicomplexan T. gondii and to identify the key components of the prenylated proteome that are involved. By exploiting the genetic tractability of T. gondii and a host GFP acquisition assay, we reveal four non-classical endocytic proteins that regulate the transport of endocytosed cargos (e.g., GFP) in T. gondii. Thus, we extend the principle that protein prenylation regulates endocytic trafficking and elucidate the process of non-classical endocytosis in T. gondii and potentially in other related protists.
Collapse
Affiliation(s)
- Qiang-Qiang Wang
- National Key Laboratory of Veterinary Public Health Security and College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Ming Sun
- National Key Laboratory of Veterinary Public Health Security and College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Tao Tang
- National Key Laboratory of Veterinary Public Health Security and College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, China
| | - De-Hua Lai
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jing Liu
- National Key Laboratory of Veterinary Public Health Security and College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Sanjay Maity
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kai He
- National Key Laboratory of Veterinary Public Health Security and College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xi-Ting Wu
- National Key Laboratory of Veterinary Public Health Security and College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jiong Yang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yue-Bao Li
- National Key Laboratory of Veterinary Public Health Security and College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiao-Yan Tang
- National Key Laboratory of Veterinary Public Health Security and College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Hui-Yong Ding
- National Key Laboratory of Veterinary Public Health Security and College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Geoff Hide
- Biomedical Research and Innovation Centre and Environmental Research and Innovation Centre, School of Science, Engineering and Environment, University of Salford, Salford, United Kingdom
| | - Mark Distefano
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota, USA
| | - Zhao-Rong Lun
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xing-Quan Zhu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi Province, China
| | - Shaojun Long
- National Key Laboratory of Veterinary Public Health Security and College of Veterinary Medicine, China Agricultural University, Beijing, China
- National Animal Protozoa Laboratory and School of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
38
|
Baba M, Nozaki M, Tachibana M, Tsuboi T, Torii M, Ishino T. Rhoptry neck protein 4 plays important roles during Plasmodium sporozoite infection of the mammalian liver. mSphere 2023; 8:e0058722. [PMID: 37272704 PMCID: PMC10449513 DOI: 10.1128/msphere.00587-22] [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: 11/20/2022] [Accepted: 04/05/2023] [Indexed: 06/06/2023] Open
Abstract
During invasion, Plasmodium parasites secrete proteins from rhoptry and microneme apical end organelles, which have crucial roles in attaching to and invading target cells. A sporozoite stage-specific gene silencing system revealed that rhoptry neck protein 2 (RON2), RON4, and RON5 are important for sporozoite invasion of mosquito salivary glands. Here, we further investigated the roles of RON4 during sporozoite infection of the liver in vivo. Following intravenous inoculation of RON4-knockdown sporozoites into mice, we demonstrated that sporozoite RON4 has multiple functions during sporozoite traversal of sinusoidal cells and infection of hepatocytes. In vitro infection experiments using a hepatoma cell line revealed that secreted RON4 is involved in sporozoite adhesion to hepatocytes and has an important role in the early steps of hepatocyte infection. In addition, in vitro motility assays indicated that RON4 is required for sporozoite attachment to the substrate and the onset of migration. These findings indicate that RON4 is crucial for sporozoite migration toward and invasion of hepatocytes via attachment ability and motility.IMPORTANCEMalarial parasite transmission to mammals is established when sporozoites are inoculated by mosquitoes and migrate through the bloodstream to infect hepatocytes. Many aspects of the molecular mechanisms underpinning migration and cellular invasion remain largely unelucidated. By applying a sporozoite stage-specific gene silencing system in the rodent malarial parasite, Plasmodium berghei, we demonstrated that rhoptry neck protein 4 (RON4) is crucial for sporozoite infection of the liver in vivo. Combined with in vitro investigations, it was revealed that RON4 functions during a crossing of the sinusoidal cell layer and invading hepatocytes, at an early stage of liver infection, by mediating the sporozoite capacity for adhesion and the onset of motility. Since RON4 is also expressed in Plasmodium merozoites and Toxoplasma tachyzoites, our findings contribute to understanding the conserved invasion mechanisms of Apicomplexa parasites.
Collapse
Affiliation(s)
- Minami Baba
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime, Japan
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
| | - Mamoru Nozaki
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime, Japan
| | - Mayumi Tachibana
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime, Japan
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime, Japan
| | - Motomi Torii
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime, Japan
| | - Tomoko Ishino
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime, Japan
- Department of Parasitology and Tropical Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
39
|
Collier S, Pietsch E, Dans M, Ling D, Tavella TA, Lopaticki S, Marapana DS, Shibu MA, Andrew D, Tiash S, McMillan PJ, Gilson P, Tilley L, Dixon MWA. Plasmodium falciparum formins are essential for invasion and sexual stage development. Commun Biol 2023; 6:861. [PMID: 37596377 PMCID: PMC10439200 DOI: 10.1038/s42003-023-05233-y] [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: 11/07/2022] [Accepted: 08/09/2023] [Indexed: 08/20/2023] Open
Abstract
The malaria parasite uses actin-based mechanisms throughout its lifecycle to control a range of biological processes including intracellular trafficking, gene regulation, parasite motility and invasion. In this work we assign functions to the Plasmodium falciparum formins 1 and 2 (FRM1 and FRM2) proteins in asexual and sexual blood stage development. We show that FRM1 is essential for merozoite invasion and FRM2 is required for efficient cell division. We also observed divergent functions for FRM1 and FRM2 in gametocyte development. Conditional deletion of FRM1 leads to a delay in gametocyte stage progression. We show that FRM2 controls the actin and microtubule cytoskeletons in developing gametocytes, with premature removal of the protein resulting in a loss of transmissible stage V gametocytes. Lastly, we show that targeting formin proteins with the small molecule inhibitor of formin homology domain 2 (SMIFH2) leads to a multistage block in asexual and sexual stage parasite development.
Collapse
Affiliation(s)
- Sophie Collier
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Emma Pietsch
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Madeline Dans
- The Macfarlane Burnet Institute for Medical Research, 85 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Dawson Ling
- The Macfarlane Burnet Institute for Medical Research, 85 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Tatyana A Tavella
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Sash Lopaticki
- Department of Infectious Diseases, Doherty Institute, University of Melbourne, Parkville, VIC, 3010, Australia
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia
| | - Danushka S Marapana
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia
| | - Mohini A Shibu
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Dean Andrew
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Snigdha Tiash
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Paul J McMillan
- Biological Optical Microscopy Platform, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Paul Gilson
- The Macfarlane Burnet Institute for Medical Research, 85 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Leann Tilley
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Matthew W A Dixon
- Department of Infectious Diseases, Doherty Institute, University of Melbourne, Parkville, VIC, 3010, Australia.
- Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC, 3052, Australia.
| |
Collapse
|
40
|
Ribeiro E. Silva A, Diallo MA, Sausset A, Robert T, Bach S, Bussière FI, Laurent F, Lacroix-Lamandé S, Silvestre A. Overexpression of Eimeria tenella Rhoptry Kinase 2 Induces Early Production of Schizonts. Microbiol Spectr 2023; 11:e0013723. [PMID: 37260371 PMCID: PMC10434272 DOI: 10.1128/spectrum.00137-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/04/2023] [Indexed: 06/02/2023] Open
Abstract
Eimeria tenella is an obligate intracellular parasite responsible for avian coccidiosis. Like other apicomplexan parasites, such as Toxoplasma gondii, cell invasion and intracellular development rely on apical organelle content discharge, named micronemes and rhoptries. Some rhoptry (ROP) kinases (ROPK) are key virulence factors in T. gondii. To date, among the 28 ropk genes carried by E. tenella, only two to four were confirmed by proteomic analysis or immunostaining to be expressed at the sporozoite stage. We have previously shown that EtROP1 is implicated in the inhibition of host cell apoptosis by interacting with the cellular p53. This work functionally described the second ROP kinase expressed at the sporozoite stage in E. tenella. EtROP2 is an active kinase that phosphorylates cell substrates of approximately 50 kDa. Its overexpression leads to the shortening of the prepatent period and to the early development of first-generation schizonts. Conduction of RNA sequencing analysis and reverse transcriptase quantitative PCR (RT-qPCR) on the host cell allowed us to identify the mitogen-activated protein kinase (MAPK) pathway and the transcription factor cFos to be upregulated by EtROP2. We also showed by immunofluorescence assay that the active kinase EtROP2 is implicated in the p38 MAPK pathway activation. We established here that EtROP2 activates the p38 MAPK pathway through a direct or indirect phosphorylation, leading to the overexpression of the master transcription factor cFos known to be implicated in E. tenella development. IMPORTANCE Rhoptries are specialized secretory organelles found in zoite stages of apicomplexan parasites. In addition to well-conserved rhoptry neck proteins, their protein consists mostly of kinase proteins, highly divergent from eukaryotic kinases. Some of those kinases are described as major virulence factors in Toxoplasma gondii, secreted into the host cell to hijack signaling pathways. Most of those kinases remain to be characterized in Eimeria tenella. Deciphering their cellular function is a prerequisite to supporting their relevance as a druggable target in development of new means of Eimeria tenella control. Secreted divergent kinases that interact with host cell partners to modulate pathways are good candidates, as they coevolve with their host targets to ensure their function within the host and are less prone to mutations that would lead to drug resistance. The absence of any orthologous kinase in host cells makes these parasite kinases a promising drug target candidate.
Collapse
Affiliation(s)
| | | | - Alix Sausset
- ISP, INRAE, Université de Tours, Nouzilly, France
| | - Thomas Robert
- Sorbonne Université, CNRS, UMR 8227, Integrative Biology of Marine Models Laboratory (LBI2M), Station Biologique de Roscoff, Roscoff, France
- Sorbonne Université, CNRS, FR 2424, Plateforme de Criblage KISSf (Kinase Inhibitor Specialized Screening Facility), Station Biologique de Roscoff, Roscoff, France
| | - Stéphane Bach
- Sorbonne Université, CNRS, UMR 8227, Integrative Biology of Marine Models Laboratory (LBI2M), Station Biologique de Roscoff, Roscoff, France
- Sorbonne Université, CNRS, FR 2424, Plateforme de Criblage KISSf (Kinase Inhibitor Specialized Screening Facility), Station Biologique de Roscoff, Roscoff, France
- Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom, South Africa
| | | | | | | | | |
Collapse
|
41
|
Back PS, Senthilkumar V, Choi CP, Ly AM, Snyder AK, Lau JG, Ward GE, Bradley PJ. The Toxoplasma subpellicular network is highly interconnected and defines parasite shape for efficient motility and replication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.10.552545. [PMID: 37609316 PMCID: PMC10441382 DOI: 10.1101/2023.08.10.552545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Apicomplexan parasites possess several specialized structures to invade their host cells and replicate successfully. One of these is the inner membrane complex (IMC), a peripheral membrane-cytoskeletal system underneath the plasma membrane. It is composed of a series of flattened, membrane-bound vesicles and a cytoskeletal subpellicular network (SPN) comprised of intermediate filament-like proteins called alveolins. While the alveolin proteins are conserved throughout the Apicomplexa and the broader Alveolata, their precise functions and interactions remain poorly understood. Here, we describe the function of one of these alveolin proteins, TgIMC6. Disruption of IMC6 resulted in striking morphological defects that led to aberrant motility, invasion, and replication. Deletion analyses revealed that the alveolin domain alone is largely sufficient to restore localization and partially sufficient for function. As this highlights the importance of the IMC6 alveolin domain, we implemented unnatural amino acid photoreactive crosslinking to the alveolin domain and identified multiple binding interfaces between IMC6 and two other cytoskeletal proteins - IMC3 and ILP1. To our knowledge, this provides the first direct evidence of protein-protein interactions in the alveolin domain and supports the long-held hypothesis that the alveolin domain is responsible for filament formation. Collectively, our study features the conserved alveolin proteins as critical components that maintain the parasite's structural integrity and highlights the alveolin domain as a key mediator of SPN architecture.
Collapse
|
42
|
Martinez M, Mageswaran SK, Guérin A, Chen WD, Thompson CP, Chavin S, Soldati-Favre D, Striepen B, Chang YW. Origin and arrangement of actin filaments for gliding motility in apicomplexan parasites revealed by cryo-electron tomography. Nat Commun 2023; 14:4800. [PMID: 37558667 PMCID: PMC10412601 DOI: 10.1038/s41467-023-40520-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 07/26/2023] [Indexed: 08/11/2023] Open
Abstract
The phylum Apicomplexa comprises important eukaryotic parasites that invade host tissues and cells using a unique mechanism of gliding motility. Gliding is powered by actomyosin motors that translocate host-attached surface adhesins along the parasite cell body. Actin filaments (F-actin) generated by Formin1 play a central role in this critical parasitic activity. However, their subcellular origin, path and ultrastructural arrangement are poorly understood. Here we used cryo-electron tomography to image motile Cryptosporidium parvum sporozoites and reveal the cellular architecture of F-actin at nanometer-scale resolution. We demonstrate that F-actin nucleates at the apically positioned preconoidal rings and is channeled into the pellicular space between the parasite plasma membrane and the inner membrane complex in a conoid extrusion-dependent manner. Within the pellicular space, filaments on the inner membrane complex surface appear to guide the apico-basal flux of F-actin. F-actin concordantly accumulates at the basal end of the parasite. Finally, analyzing a Formin1-depleted Toxoplasma gondii mutant pinpoints the upper preconoidal ring as the conserved nucleation hub for F-actin in Cryptosporidium and Toxoplasma. Together, we provide an ultrastructural model for the life cycle of F-actin for apicomplexan gliding motility.
Collapse
Affiliation(s)
- Matthew Martinez
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shrawan Kumar Mageswaran
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute of Structural Biology, 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
| | - William David Chen
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cameron Parker Thompson
- Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sabine Chavin
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA, USA
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - 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.
- Institute of Structural Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
43
|
Tremp AZ, Saeed S, Dessens JT. NAD(P) transhydrogenase isoform distribution provides insight into apicomplexan evolution. Front Ecol Evol 2023; 11:fevo.2023.1216385. [PMID: 37581102 PMCID: PMC7614955 DOI: 10.3389/fevo.2023.1216385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2023] Open
Abstract
Membrane-located NAD(P) transhydrogenase (NTH) catalyses reversible hydride ion transfer between NAD(H) and NADP(H), simultaneously translocating a proton across the membrane. The enzyme is structurally conserved across prokaryotes and eukaryotes. In heterotrophic bacteria NTH proteins reside in the cytoplasmic membrane, whereas in animals they localise in the mitochondrial inner membrane. Eukaryotic NTH proteins exists in two distinct configurations (isoforms) and have non-mitochondrial functions in unicellular eukaryotes like Plasmodium, the causative agent of malaria. In this study, we carried out a systematic analysis of nth genes across eukaryotic life to determine its prevalence and distribution of isoforms. The results reveal that NTH is found across all major lineages, but that some organisms, notably plants, lack nth genes altogether. Isoform distribution and phylogenetic analysis reveals different nth gene loss scenarios in apicomplexan lineages, which sheds new light on the evolution of the Piroplasmida and Eimeriidae.
Collapse
Affiliation(s)
- Annie Z. Tremp
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Sadia Saeed
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Johannes T. Dessens
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| |
Collapse
|
44
|
He L, Qiu Y, Pang G, Li S, Wang J, Feng Y, Chen L, Zhu L, Liu Y, Cui L, Cao Y, Zhu X. Plasmodium falciparum GAP40 Plays an Essential Role in Merozoite Invasion and Gametocytogenesis. Microbiol Spectr 2023; 11:e0143423. [PMID: 37249423 PMCID: PMC10269477 DOI: 10.1128/spectrum.01434-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/03/2023] [Indexed: 05/31/2023] Open
Abstract
Cyclic invasion of red blood cells (RBCs) by Plasmodium merozoites is associated with the symptoms and pathology of malaria. Merozoite invasion is powered actively and rapidly by a parasite actomyosin motor called the glideosome. The ability of the glideosome to generate force to support merozoite entry into the host RBCs is thought to rely on its stable anchoring within the inner membrane complex (IMC) through membrane-resident proteins, such as GAP50 and GAP40. Using a conditional knockdown (KD) approach, we determined that PfGAP40 was required for asexual blood-stage replication. PfGAP40 is not needed for merozoite egress from host RBCs or for the attachment of merozoites to new RBCs. PfGAP40 coprecipitates with PfGAP45 and PfGAP50. During merozoite invasion, PfGAP40 is associated strongly with stabilizing the expression levels of PfGAP45 and PfGAP50 in the schizont stage. Although PfGAP40 KD did not influence IMC integrity, it impaired the maturation of gametocytes. In addition, PfGAP40 is phosphorylated, and mutations that block phosphorylation of PfGAP40 at the C-terminal serine residues S370, S372, S376, S405, S409, S420, and S445 reduced merozoite invasion efficiency. Overall, our findings implicate PfGAP40 as an important regulator for the gliding activity of merozoites and suggest that phosphorylation is required for PfGAP40 function. IMPORTANCE Red blood cell invasion is central to the pathogenesis of the malaria parasite, and the parasite proteins involved in this process are potential therapeutic targets. Gliding motility powers merozoite invasion and is driven by a unique molecular motor termed the glideosome. The glideosome is stably anchored to the parasite inner membrane complex (IMC) through membrane-resident proteins. In the present study, we demonstrate the importance of an IMC-resident glideosome component, PfGAP40, that plays a critical role in stabilizing the expression levels of glideosome components in the schizont stage. We determined that phosphorylation of PfGAP40 at C-terminal residues is required for efficient merozoite invasion.
Collapse
Affiliation(s)
- Lu He
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Yue Qiu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Geping Pang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Siqi Li
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Jingjing Wang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Yonghui Feng
- Department of Laboratory Medicine, the First Hospital of China Medical University, Shenyang, Liaoning, China
- National Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning, China
| | - Lumeng Chen
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Liying Zhu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Yinjie Liu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Liwang Cui
- College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - Xiaotong Zhu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
45
|
Moussaoui D, Robblee JP, Robert-Paganin J, Auguin D, Fisher F, Fagnant PM, Macfarlane JE, Schaletzky J, Wehri E, Mueller-Dieckmann C, Baum J, Trybus KM, Houdusse A. Mechanism of small molecule inhibition of Plasmodium falciparum myosin A informs antimalarial drug design. Nat Commun 2023; 14:3463. [PMID: 37308472 PMCID: PMC10261046 DOI: 10.1038/s41467-023-38976-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 05/24/2023] [Indexed: 06/14/2023] Open
Abstract
Malaria results in more than 500,000 deaths per year and the causative Plasmodium parasites continue to develop resistance to all known agents, including different antimalarial combinations. The class XIV myosin motor PfMyoA is part of a core macromolecular complex called the glideosome, essential for Plasmodium parasite mobility and therefore an attractive drug target. Here, we characterize the interaction of a small molecule (KNX-002) with PfMyoA. KNX-002 inhibits PfMyoA ATPase activity in vitro and blocks asexual blood stage growth of merozoites, one of three motile Plasmodium life-cycle stages. Combining biochemical assays and X-ray crystallography, we demonstrate that KNX-002 inhibits PfMyoA using a previously undescribed binding mode, sequestering it in a post-rigor state detached from actin. KNX-002 binding prevents efficient ATP hydrolysis and priming of the lever arm, thus inhibiting motor activity. This small-molecule inhibitor of PfMyoA paves the way for the development of alternative antimalarial treatments.
Collapse
Affiliation(s)
- Dihia Moussaoui
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, 75248, Paris, France
- Structural Biology group, European Synchrotron Radiation Facility (ESRF), 71, Avenue des Martyrs, 38000, Grenoble, France
| | - James P Robblee
- Department of Molecular Physiology & Biophysics, University of Vermont, Burlington, VT, USA
| | - Julien Robert-Paganin
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, 75248, Paris, France
| | - Daniel Auguin
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, 75248, Paris, France
- Laboratoire de Biologie des Ligneux et des Grandes Cultures (LBLGC), Université d'Orléans, INRAE, USC1328, Orléans, France
| | - Fabio Fisher
- Department of Life Sciences, Imperial College London, Exhibition Road, South Kensington, London, SW7 2AZ, UK
| | - Patricia M Fagnant
- Department of Molecular Physiology & Biophysics, University of Vermont, Burlington, VT, USA
| | - Jill E Macfarlane
- Department of Molecular Physiology & Biophysics, University of Vermont, Burlington, VT, USA
| | - Julia Schaletzky
- Center for Emerging and Neglected Diseases, Drug Discovery Center, Berkeley, CA, USA
| | - Eddie Wehri
- Center for Emerging and Neglected Diseases, Drug Discovery Center, Berkeley, CA, USA
| | - Christoph Mueller-Dieckmann
- Structural Biology group, European Synchrotron Radiation Facility (ESRF), 71, Avenue des Martyrs, 38000, Grenoble, France
| | - Jake Baum
- Department of Life Sciences, Imperial College London, Exhibition Road, South Kensington, London, SW7 2AZ, UK
- School of Medical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, NSW, 2052, Australia
| | - Kathleen M Trybus
- Department of Molecular Physiology & Biophysics, University of Vermont, Burlington, VT, USA.
| | - Anne Houdusse
- Structural Motility, Institut Curie, Université Paris Sciences et Lettres, Sorbonne Université, CNRS UMR144, 75248, Paris, France.
| |
Collapse
|
46
|
Zhang Z, Gaetjens TK, Yu Y, Paul Mallory D, Abel SM, Yu Y. Propulsive cell entry diverts pathogens from immune degradation by remodeling the phagocytic synapse. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.25.538287. [PMID: 37162866 PMCID: PMC10168248 DOI: 10.1101/2023.04.25.538287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Phagocytosis is a critical immune function for infection control and tissue homeostasis. This process is typically described as non-moving pathogens being internalized and degraded in phagolysosomes. For pathogens that evade immune degradation, the prevailing view is that virulence factors that biochemically disrupt the biogenesis of phagoslysosomes are required. In contrast, here we report that physical forces exerted by pathogens during cell entry divert them away from the canonical phagolysosomal degradation pathway, and this altered intracellular fate is determined at the time of phagocytic synapse formation. We used the eukaryotic parasite Toxoplasma gondii as a model because live Toxoplasma uses gliding motility to actively invade into host cells. To differentiate the effect of physical forces from that of virulence factors in phagocytosis, we developed a strategy that used magnetic forces to induce propulsive entry of inactivated Toxoplasma into macrophage cells. Experiments and computer simulations collectively reveal that large propulsive forces suppress productive activation of receptors by hindering their spatial segregation from phosphatases at the phagocytic synapse. Consequently, the inactivated parasites, instead of being degraded in phagolysosomes, are engulfed into vacuoles that fail to mature into degradative units, following an intracellular pathway strikingly similar to that of the live motile parasite. Using opsonized beads, we further confirmed that this mechanism is general, not specific to the parasite used. These results reveal previously unknown aspects of immune evasion by demonstrating how physical forces exerted during active cell entry, independent of virulence factors, can help pathogens circumvent phagolysosomal degradation.
Collapse
Affiliation(s)
- Zihan Zhang
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102
| | - Thomas K. Gaetjens
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996
| | - Yanqi Yu
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102
| | - D. Paul Mallory
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102
| | - Steven M. Abel
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996
| | - Yan Yu
- Department of Chemistry, Indiana University, Bloomington, IN 47405-7102
| |
Collapse
|
47
|
Koreny L, Mercado-Saavedra BN, Klinger CM, Barylyuk K, Butterworth S, Hirst J, Rivera-Cuevas Y, Zaccai NR, Holzer VJC, Klingl A, Dacks JB, Carruthers VB, Robinson MS, Gras S, Waller RF. Stable endocytic structures navigate the complex pellicle of apicomplexan parasites. Nat Commun 2023; 14:2167. [PMID: 37061511 PMCID: PMC10105704 DOI: 10.1038/s41467-023-37431-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 03/17/2023] [Indexed: 04/17/2023] Open
Abstract
Apicomplexan parasites have immense impacts on humanity, but their basic cellular processes are often poorly understood. Where endocytosis occurs in these cells, how conserved this process is with other eukaryotes, and what the functions of endocytosis are across this phylum are major unanswered questions. Using the apicomplexan model Toxoplasma, we identified the molecular composition and behavior of unusual, fixed endocytic structures. Here, stable complexes of endocytic proteins differ markedly from the dynamic assembly/disassembly of these machineries in other eukaryotes. We identify that these endocytic structures correspond to the 'micropore' that has been observed throughout the Apicomplexa. Moreover, conserved molecular adaptation of this structure is seen in apicomplexans including the kelch-domain protein K13 that is central to malarial drug-resistance. We determine that a dominant function of endocytosis in Toxoplasma is plasma membrane homeostasis, rather than parasite nutrition, and that these specialized endocytic structures originated early in infrakingdom Alveolata likely in response to the complex cell pellicle that defines this medically and ecologically important ancient eukaryotic lineage.
Collapse
Affiliation(s)
- Ludek Koreny
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QW, UK
| | | | - Christen M Klinger
- Division of Infectious Diseases, Department of Medicine, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | | | - Simon Butterworth
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QW, UK
| | - Jennifer Hirst
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 1QW, UK
| | - Yolanda Rivera-Cuevas
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Nathan R Zaccai
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 1QW, UK
| | - Victoria J C Holzer
- Plant Development, Ludwig-Maximilians-University Munich, Planegg-Martinsried, 82152, Germany
| | - Andreas Klingl
- Plant Development, Ludwig-Maximilians-University Munich, Planegg-Martinsried, 82152, Germany
| | - Joel B Dacks
- Division of Infectious Diseases, Department of Medicine, University of Alberta, Edmonton, AB, T6G 2R3, Canada
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice, 370 05, Czech Republic
| | - Vern B Carruthers
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Margaret S Robinson
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 1QW, UK
| | - Simon Gras
- Experimental Parasitology, Department for Veterinary Sciences, Ludwig-Maximilians-University Munich, Planegg-Martinsried, 82152, Germany.
| | - Ross F Waller
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QW, UK.
| |
Collapse
|
48
|
Kumar A, Vadas O, Dos Santos Pacheco N, Zhang X, Chao K, Darvill N, Rasmussen HØ, Xu Y, Lin GMH, Stylianou FA, Pedersen JS, Rouse SL, Morgan ML, Soldati-Favre D, Matthews S. Structural and regulatory insights into the glideosome-associated connector from Toxoplasma gondii. eLife 2023; 12:e86049. [PMID: 37014051 PMCID: PMC10125020 DOI: 10.7554/elife.86049] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/03/2023] [Indexed: 04/05/2023] Open
Abstract
The phylum of Apicomplexa groups intracellular parasites that employ substrate-dependent gliding motility to invade host cells, egress from the infected cells, and cross biological barriers. The glideosome-associated connector (GAC) is a conserved protein essential to this process. GAC facilitates the association of actin filaments with surface transmembrane adhesins and the efficient transmission of the force generated by myosin translocation of actin to the cell surface substrate. Here, we present the crystal structure of Toxoplasma gondii GAC and reveal a unique, supercoiled armadillo repeat region that adopts a closed ring conformation. Characterisation of the solution properties together with membrane and F-actin binding interfaces suggests that GAC adopts several conformations from closed to open and extended. A multi-conformational model for assembly and regulation of GAC within the glideosome is proposed.
Collapse
Affiliation(s)
- Amit Kumar
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Oscar Vadas
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of GenevaGenevaSwitzerland
| | - Nicolas Dos Santos Pacheco
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of GenevaGenevaSwitzerland
| | - Xu Zhang
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Kin Chao
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Nicolas Darvill
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Helena Ø Rasmussen
- Interdisciplinary Nanoscience Center (iNANO) and Department of Chemistry, Aarhus UniversityAarhusDenmark
| | - Yingqi Xu
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Gloria Meng-Hsuan Lin
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of GenevaGenevaSwitzerland
| | | | - Jan Skov Pedersen
- Interdisciplinary Nanoscience Center (iNANO) and Department of Chemistry, Aarhus UniversityAarhusDenmark
| | - Sarah L Rouse
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Marc L Morgan
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of GenevaGenevaSwitzerland
| | - Stephen Matthews
- Department of Life Sciences, Imperial College LondonLondonUnited Kingdom
| |
Collapse
|
49
|
Ong HW, Adderley J, Tobin AB, Drewry DH, Doerig C. Parasite and host kinases as targets for antimalarials. Expert Opin Ther Targets 2023; 27:151-169. [PMID: 36942408 DOI: 10.1080/14728222.2023.2185511] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
INTRODUCTION The deployment of Artemisinin-based combination therapies and transmission control measures led to a decrease in the global malaria burden over the recent decades. Unfortunately, this trend is now reversing, in part due to resistance against available treatments, calling for the development of new drugs against untapped targets to prevent cross-resistance. AREAS COVERED In view of their demonstrated druggability in noninfectious diseases, protein kinases represent attractive targets. Kinase-focussed antimalarial drug discovery is facilitated by the availability of kinase-targeting scaffolds and large libraries of inhibitors, as well as high-throughput phenotypic and biochemical assays. We present an overview of validated Plasmodium kinase targets and their inhibitors, and briefly discuss the potential of host cell kinases as targets for host-directed therapy. EXPERT OPINION We propose priority research areas, including (i) diversification of Plasmodium kinase targets (at present most efforts focus on a very small number of targets); (ii) polypharmacology as an avenue to limit resistance (kinase inhibitors are highly suitable in this respect); and (iii) preemptive limitation of resistance through host-directed therapy (targeting host cell kinases that are required for parasite survival) and transmission-blocking through targeting sexual stage-specific kinases as a strategy to protect curative drugs from the spread of resistance.
Collapse
Affiliation(s)
- Han Wee Ong
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC USA
| | - Jack Adderley
- Department of Laboratory Medicine, School of Health and Biomedical Sciences, Rmit University, Bundoora VIC Australia
| | - Andrew B Tobin
- Advanced Research Centre, University of Glasgow, Glasgow, UK
| | - David H Drewry
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC USA
| | - Christian Doerig
- Department of Laboratory Medicine, School of Health and Biomedical Sciences, Rmit University, Bundoora VIC Australia
| |
Collapse
|
50
|
Guérin A, Strelau KM, Barylyuk K, Wallbank BA, Berry L, Crook OM, Lilley KS, Waller RF, Striepen B. Cryptosporidium uses multiple distinct secretory organelles to interact with and modify its host cell. Cell Host Microbe 2023; 31:650-664.e6. [PMID: 36958336 DOI: 10.1016/j.chom.2023.03.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/09/2023] [Accepted: 02/28/2023] [Indexed: 03/25/2023]
Abstract
Cryptosporidium is a leading cause of diarrheal disease in children and an important contributor to early childhood mortality. The parasite invades and extensively remodels intestinal epithelial cells, building an elaborate interface structure. How this occurs at the molecular level and the contributing parasite factors are largely unknown. Here, we generated a whole-cell spatial proteome of the Cryptosporidium sporozoite and used genetic and cell biological experimentation to discover the Cryptosporidium-secreted effector proteome. These findings reveal multiple organelles, including an original secretory organelle, and generate numerous compartment markers by tagging native gene loci. We show that secreted proteins are delivered to the parasite-host interface, where they assemble into different structures including a ring that anchors the parasite into its unique epicellular niche. Cryptosporidium thus uses a complex set of secretion systems during and following invasion that act in concert to subjugate its host cell.
Collapse
Affiliation(s)
- Amandine Guérin
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Katherine M Strelau
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Bethan A Wallbank
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Laurence Berry
- LPHI, CNRS, Université de Montpellier, Montpellier 34095, France
| | - Oliver M Crook
- Department of Statistics, University of Oxford, Oxford OX1 3LB, UK
| | - Kathryn S Lilley
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Ross F Waller
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|