1
|
Ghosh A, Mishra A, Devi R, Narwal SK, Nirdosh, Srivastava PN, Mishra S. A Micronemal Protein, Scot1, Is Essential for Apicoplast Biogenesis and Liver Stage Development in Plasmodium berghei. ACS Infect Dis 2024; 10:3013-3025. [PMID: 39037752 DOI: 10.1021/acsinfecdis.4c00362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Plasmodium sporozoites invade hepatocytes, transform into liver stages, and replicate into thousands of merozoites that infect erythrocytes and cause malaria. Proteins secreted from micronemes play an essential role in hepatocyte invasion, and unneeded micronemes are subsequently discarded for replication. The liver-stage parasites are potent immunogens that prevent malarial infection. Late liver stage-arresting genetically attenuated parasites (GAPs) exhibit greater protective efficacy than early GAP. However, the number of late liver-stage GAPs for generating GAPs with multiple gene deletions is limited. Here, we identified Scot1 (Sporozoite Conserved Orthologous Transcript 1), which was previously shown to be upregulated in sporozoites, and by endogenous tagging with mCherry, we demonstrated that it is expressed in the sporozoite and liver stages in micronemes. Using targeted gene deletion in Plasmodium berghei, we showed that Scot1 is essential for late liver-stage development. Scot1 KO sporozoites grew normally into liver stages but failed to initiate blood-stage infection in mice due to impaired apicoplast biogenesis and merozoite formation. Bioinformatic studies suggested that Scot1 is a metal-small-molecule carrier protein. Remarkably, supplementation with metals in the culture of infected Scot1 KO cells did not rescue their phenotype. Immunization with Scot1 KO sporozoites in C57BL/6 mice confers protection against malaria via infection. These proof-of-concept studies will enable the generation of P. falciparum Scot1 mutants that could be exploited to generate GAP malaria vaccines.
Collapse
Affiliation(s)
- Ankit Ghosh
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Akancha 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
| | - Raksha Devi
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, 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
| | - Pratik Narain Srivastava
- 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
|
2
|
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
|
3
|
Segireddy RR, Belda H, Yang ASP, Dundas K, Knoeckel J, Galaway F, Wood L, Quinkert D, Knuepfer E, Treeck M, Wright GJ, Douglas AD. A screen for Plasmodium falciparum sporozoite surface protein binding to human hepatocyte surface receptors identifies novel host-pathogen interactions. Malar J 2024; 23:151. [PMID: 38755636 PMCID: PMC11098746 DOI: 10.1186/s12936-024-04913-2] [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: 12/07/2023] [Accepted: 03/20/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Sporozoite invasion of hepatocytes is an essential step in the Plasmodium life-cycle and has similarities, at the cellular level, to merozoite invasion of erythrocytes. In the case of the Plasmodium blood-stage, efforts to identify host-pathogen protein-protein interactions have yielded important insights including vaccine candidates. In the case of sporozoite-hepatocyte invasion, the host-pathogen protein-protein interactions involved are poorly understood. METHODS To gain a better understanding of the protein-protein interaction between the sporozoite ligands and host receptors, a systematic screen was performed. The previous Plasmodium falciparum and human surface protein ectodomain libraries were substantially extended, resulting in the creation of new libraries comprising 88 P. falciparum sporozoite protein coding sequences and 182 sequences encoding human hepatocyte surface proteins. Having expressed recombinant proteins from these sequences, a plate-based assay was used, capable of detecting low affinity interactions between recombinant proteins, modified for enhanced throughput, to screen the proteins for interactions. The novel interactions identified in the screen were characterized biochemically, and their essential role in parasite invasion was further elucidated using antibodies and genetically manipulated Plasmodium parasites. RESULTS A total of 7540 sporozoite-hepatocyte protein pairs were tested under conditions capable of detecting interactions of at least 1.2 µM KD. An interaction between the human fibroblast growth factor receptor 4 (FGFR4) and the P. falciparum protein Pf34 is identified and reported here, characterizing its affinity and demonstrating the blockade of the interaction by reagents, including a monoclonal antibody. Furthermore, further interactions between Pf34 and a second P. falciparum rhoptry neck protein, PfRON6, and between human low-density lipoprotein receptor (LDLR) and the P. falciparum protein PIESP15 are identified. Conditional genetic deletion confirmed the essentiality of PfRON6 in the blood-stage, consistent with the important role of this protein in parasite lifecycle. Pf34 was refractory to attempted genetic modification. Antibodies to Pf34 abrogated the interaction and had a modest effect upon sporozoite invasion into primary human hepatocytes. CONCLUSION Pf34 and PfRON6 may be members of a functionally important invasion complex which could be a target for future interventions. The modified interaction screening assay, protein expression libraries and P. falciparum mutant parasites reported here may be a useful tool for protein interaction discovery and antigen candidate screening which could be of wider value to the scientific community.
Collapse
Affiliation(s)
- Rameswara R Segireddy
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK.
| | - Hugo Belda
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick Institute, Midland Road, London, NW1 1AT, UK
| | - Annie S P Yang
- Research Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Kirsten Dundas
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Julia Knoeckel
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Francis Galaway
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Laura Wood
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Doris Quinkert
- Department of Biochemistry, University of Oxford, South Parks Rd, Oxford, OX1 3QU, UK
| | - Ellen Knuepfer
- The Royal Veterinary College, North Mymms, Hawkshead Lane, Hatfield, Hertfordshire, AL9 7TA, UK
| | - Moritz Treeck
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick Institute, Midland Road, London, NW1 1AT, UK
| | - Gavin J Wright
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York, Wentworth Way, York, YO10 5DD, UK
| | - Alexander D Douglas
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK.
| |
Collapse
|
4
|
Glennon EK, Wei L, Roobsoong W, Primavera VI, Tongogara T, Yee CB, Sattabongkot J, Kaushansky A. Host kinase regulation of Plasmodium vivax dormant and replicating liver stages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.13.566868. [PMID: 38014051 PMCID: PMC10680662 DOI: 10.1101/2023.11.13.566868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Upon transmission to the liver, Plasmodium vivax parasites form replicating schizonts, which continue to initiate blood-stage infection, or dormant hypnozoites that reactivate weeks to months after initial infection. P. vivax phenotypes in the field vary significantly, including the ratio of schizonts to hypnozoites formed and the frequency and timing of relapse. Evidence suggests that both parasite genetics and environmental factors underly this heterogeneity. We previously demonstrated that data on the effect of a panel of kinase inhibitors with overlapping targets on Plasmodium liver stage infection, in combination with a computational approach called kinase regression (KiR), can be used to uncover novel host regulators of infection. Here, we applied KiR to evaluate the extent to which P. vivax liver-stage parasites are susceptible to changes in host kinase activity. We identified a role for a subset of host kinases in regulating schizont and hypnozoite infection and schizont size and characterized overlap as well as variability in host phosphosignaling dependencies between parasite forms and across multiple patient isolates. Striking, our data point to variability in host dependencies across P. vivax isolates, suggesting one possible origin of the heterogeneity observed across P. vivax in the field.
Collapse
|
5
|
Klug D, Blandin SA. Activation of complement-like antiparasitic responses in Anopheles mosquitoes. Curr Opin Microbiol 2023; 72:102280. [PMID: 36841199 DOI: 10.1016/j.mib.2023.102280] [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: 11/21/2022] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 02/25/2023]
Abstract
During their development in mosquitoes, malaria parasites undergo massive losses that are in part due to a potent antiparasitic response mounted by the vector. The most efficient and best-characterized response relies on a complement-like system particularly effective against parasites as they cross the mosquito midgut epithelium. While our vision of the molecular and cellular events that lead to parasite elimination is still partial, our understanding of the steps triggering complement activation at the surface of invading parasites has considerably progressed, not only through the identification of novel contributing genes, but also with the recent in-depth characterization of the different mosquito blood cell types, and the ability to track them in live mosquitoes. Here, we propose a simple model based on the time of invasion to explain how parasites may escape complement-like responses during midgut infection.
Collapse
Affiliation(s)
- Dennis Klug
- Université de Strasbourg, CNRS, Inserm, UPR9022/U1257, Mosquito Immune Responses (MIR), F-67000 Strasbourg, France
| | - Stephanie A Blandin
- Université de Strasbourg, CNRS, Inserm, UPR9022/U1257, Mosquito Immune Responses (MIR), F-67000 Strasbourg, France.
| |
Collapse
|
6
|
Plasmodium 6-cysteine proteins determine the commitment of sporozoites to liver-infection. Parasitol Int 2023; 93:102700. [PMID: 36403748 DOI: 10.1016/j.parint.2022.102700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/11/2022] [Accepted: 11/13/2022] [Indexed: 11/19/2022]
Abstract
Plasmodium sporozoites travel a long way from the site where they are released by a mosquito bite to the liver, where they infect hepatocytes and develop into erythrocyte-invasive forms. The success of this infection depends on the ability of the sporozoites to correctly recognize the hepatocyte as a target and change their behavior from migration to infection. However, how this change is accomplished remains incompletely understood. In this paper, we report that 6-cysteine protein family members expressed in sporozoites including B9 are responsible for this ability. Experiments on parasites using double knockouts of B9 and SPECT2, which is essential for sporozoite to migrate through the hepatocyte, showed that the parasites lacked the capacity to stop migration. This finding suggests that interactions between these parasite proteins and hepatocyte-specific cell surface ligands mediate correct recognition of hepatocytes by sporozoites, which is an essential step in malaria transmission to humans.
Collapse
|
7
|
Loubens M, Marinach C, Paquereau CE, Hamada S, Hoareau-Coudert B, Akbar D, Franetich JF, Silvie O. The claudin-like apicomplexan microneme protein is required for gliding motility and infectivity of Plasmodium sporozoites. PLoS Pathog 2023; 19:e1011261. [PMID: 36928686 PMCID: PMC10047546 DOI: 10.1371/journal.ppat.1011261] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 03/28/2023] [Accepted: 03/02/2023] [Indexed: 03/18/2023] Open
Abstract
Invasion of host cells by apicomplexan parasites such as Toxoplasma and Plasmodium spp requires the sequential secretion of the parasite apical organelles, the micronemes and the rhoptries. The claudin-like apicomplexan microneme protein (CLAMP) is a conserved protein that plays an essential role during invasion by Toxoplasma gondii tachyzoites and in Plasmodium falciparum asexual blood stages. CLAMP is also expressed in Plasmodium sporozoites, the mosquito-transmitted forms of the malaria parasite, but its role in this stage is still unknown. CLAMP is essential for Plasmodium blood stage growth and is refractory to conventional gene deletion. To circumvent this obstacle and study the function of CLAMP in sporozoites, we used a conditional genome editing strategy based on the dimerisable Cre recombinase in the rodent malaria model parasite P. berghei. We successfully deleted clamp gene in P. berghei transmission stages and analyzed the functional consequences on sporozoite infectivity. In mosquitoes, sporozoite development and egress from oocysts was not affected in conditional mutants. However, invasion of the mosquito salivary glands was dramatically reduced upon deletion of clamp gene. In addition, CLAMP-deficient sporozoites were impaired in cell traversal and productive invasion of mammalian hepatocytes. This severe phenotype was associated with major defects in gliding motility and with reduced shedding of the sporozoite adhesin TRAP. Expansion microscopy revealed partial colocalization of CLAMP and TRAP in a subset of micronemes, and a distinct accumulation of CLAMP at the apical tip of sporozoites. Collectively, these results demonstrate that CLAMP is essential across invasive stages of the malaria parasite, and support a role of the protein upstream of host cell invasion, possibly by regulating the secretion or function of adhesins in Plasmodium sporozoites.
Collapse
Affiliation(s)
- Manon Loubens
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Carine Marinach
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Clara-Eva Paquereau
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Soumia Hamada
- Sorbonne Université, INSERM, UMS PASS, Plateforme Post-génomique de la Pitié Salpêtrière (P3S), Paris, France
| | - Bénédicte Hoareau-Coudert
- Sorbonne Université, INSERM, UMS PASS, Plateforme de cytométrie de la Pitié-Salpêtrière (CyPS), Paris, France
| | - David Akbar
- Sorbonne Université, INSERM, CNRS, Hôpital de la Pitié Salpêtrière, Paris Brain Institute, ICM Quant Cell imaging Core Facility, Paris, France
| | - Jean-François Franetich
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Olivier Silvie
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| |
Collapse
|
8
|
Karam J, Blanchet FP, Vivès É, Boisguérin P, Boudehen YM, Kremer L, Daher W. Mycobacterium abscessus alkyl hydroperoxide reductase C promotes cell invasion by binding to tetraspanin CD81. iScience 2023; 26:106042. [PMID: 36818301 PMCID: PMC9929602 DOI: 10.1016/j.isci.2023.106042] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/19/2022] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Mycobacterium abscessus (Mab) is an increasingly recognized pulmonary pathogen. How Mab is internalized by macrophages and establishes infection remains unknown. Here, we show that Mab uptake is significantly reduced in macrophages pre-incubated with neutralizing anti-CD81 antibodies or in cells in which the large extracellular loop (LEL) of CD81 has been deleted. Saturation of Mab with either soluble GST-CD81-LEL or CD81-LEL-derived peptides also diminished internalization of the bacilli. The mycobacterial alkyl hydroperoxide reductase C (AhpC) was unveiled as a major interactant of CD81-LEL. Pre-exposure of macrophages with soluble AhpC inhibited mycobacterial uptake whereas overexpression of AhpC in Mab enhanced its internalization. Importantly, pre-incubation of macrophages with anti-CD81-LEL antibodies inhibited phagocytosis of AhpC-coated beads, indicating that AhpC is a direct interactant of CD81-LEL. Conditional depletion of AhpC in Mab correlated with decreased internalization of Mab. These compelling data unravel a previously unexplored role for CD81/AhpC to promote uptake of pathogenic mycobacteria by host cells.
Collapse
Affiliation(s)
- Jona Karam
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 Route de Mende, 34293 Montpellier, France
| | - Fabien P. Blanchet
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 Route de Mende, 34293 Montpellier, France
- INSERM, IRIM, 34293 Montpellier, France
| | - Éric Vivès
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR, 9214 Montpellier, France
| | - Prisca Boisguérin
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR, 9214 Montpellier, France
| | - Yves-Marie Boudehen
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 Route de Mende, 34293 Montpellier, France
| | - Laurent Kremer
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 Route de Mende, 34293 Montpellier, France
- INSERM, IRIM, 34293 Montpellier, France
| | - Wassim Daher
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 Route de Mende, 34293 Montpellier, France
- INSERM, IRIM, 34293 Montpellier, France
| |
Collapse
|
9
|
Schepis A, Kumar S, Kappe SHI. Malaria parasites harness Rho GTPase signaling and host cell membrane ruffling for productive invasion of hepatocytes. Cell Rep 2023; 42:111927. [PMID: 36640315 DOI: 10.1016/j.celrep.2022.111927] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 09/06/2022] [Accepted: 12/14/2022] [Indexed: 12/31/2022] Open
Abstract
Plasmodium sporozoites are the motile forms of the malaria parasites that infect hepatocytes. The initial invasion of hepatocytes is thought to be actively driven by sporozoites, but host cell processes might also play a role. Sporozoite invasion triggers a host plasma membrane invagination that forms a vacuole around the intracellular parasite, which is critical for subsequent intracellular parasite replication. Using fast live confocal microscopy, we observed that the initial interactions between sporozoites and hepatocytes induce plasma membrane ruffles and filopodia extensions. Importantly, we find that these host cell processes facilitate invasion and that Rho GTPase signaling, which regulates membrane ruffling and filopodia extension, is critical for productive infection. Interestingly, sporozoite cell traversal stimulates these processes, suggesting that it increases hepatocyte susceptibility to productive infection. Our study identifies host cell signaling events involved in plasma membrane dynamics as a critical host component of successful malaria parasite infection of hepatocytes.
Collapse
Affiliation(s)
- Antonino Schepis
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Seattle, WA 98109, USA
| | - Sudhir Kumar
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Seattle, WA 98109, USA
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Seattle, WA 98109, USA; Department of Pediatrics, University of Washington, Seattle, WA 98105, USA; Department of Global Health, University of Washington, Seattle, WA 98105, USA.
| |
Collapse
|
10
|
Fernandes P, Loubens M, Marinach C, Coppée R, Baron L, Grand M, Andre TP, Hamada S, Langlois AC, Briquet S, Bun P, Silvie O. Plasmodium sporozoites require the protein B9 to invade hepatocytes. iScience 2023; 26:106056. [PMID: 36761022 PMCID: PMC9906020 DOI: 10.1016/j.isci.2023.106056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 11/16/2022] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Plasmodium sporozoites are transmitted to a mammalian host during blood feeding by an infected mosquito and invade hepatocytes for initial replication of the parasite into thousands of erythrocyte-invasive merozoites. Here we report that the B9 protein, a member of the 6-cysteine domain protein family, is secreted from sporozoite micronemes and is required for productive invasion of hepatocytes. The N-terminus of B9 forms a beta-propeller domain structurally related to CyRPA, a cysteine-rich protein forming an essential invasion complex in Plasmodium falciparum merozoites. The beta-propeller domain of B9 is essential for sporozoite infectivity and interacts with the 6-cysteine proteins P36 and P52 in a heterologous expression system. Our results suggest that, despite using distinct sets of parasite and host entry factors, Plasmodium sporozoites and merozoites may share common structural modules to assemble protein complexes for invasion of host cells.
Collapse
Affiliation(s)
- Priyanka Fernandes
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Manon Loubens
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Carine Marinach
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Romain Coppée
- Université de Paris, UMR 261 MERIT, IRD, 75006 Paris, France
| | - Ludivine Baron
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Morgane Grand
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Thanh-Phuc Andre
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Soumia Hamada
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
- Sorbonne Université, INSERM, UMS PASS, Plateforme Post-génomique de la Pitié Salpêtrière (P3S), 75013 Paris, France
| | - Anne-Claire Langlois
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Sylvie Briquet
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Philippe Bun
- INSERM U1266, NeurImag Facility, Institute of Psychiatry and Neurosciences of Paris, Paris, France
| | - Olivier Silvie
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
- Corresponding author
| |
Collapse
|
11
|
Overexpression of hepatocyte EphA2 enhances liver-stage infection by Plasmodium vivax. Sci Rep 2022; 12:21542. [PMID: 36513700 PMCID: PMC9746569 DOI: 10.1038/s41598-022-25281-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022] Open
Abstract
The liver is the first destination of malaria parasites in humans. After reaching the liver by the blood stream, Plasmodium sporozoites cross the liver sinusoid epithelium, enter and exit several hepatocytes, and eventually invade a final hepatocyte host cell. At present, the mechanism of hepatocyte invasion is only partially understood, presenting a key research gap with opportunities for the development of new therapeutics. Recently, human EphA2, a membrane-bound receptor tyrosine kinase, was implicated in hepatocyte infection by the human malaria parasite Plasmodium falciparum and the rodent parasite Plasmodium yoelii, but its role is not known for Plasmodium vivax, a major human parasite whose liver infection poses a specific challenge for malaria treatment and elimination. In this study, the role of EphA2 in P. vivax infection was investigated. It was found that surface expression of several recombinant fragments of EphA2 enhanced the parasite infection rate, thus establishing its role in P. vivax infection. Furthermore, a new permanent cell line (EphA2Extra-HC04) expressing the whole extracellular domain of EphA2 was generated. This cell line supports a higher rate of P. vivax infection and is a valuable tool for P. vivax liver-stage research.
Collapse
|
12
|
Vantaux A, Péneau J, Cooper CA, Kyle DE, Witkowski B, Maher SP. Liver-stage fate determination in Plasmodium vivax parasites: Characterization of schizont growth and hypnozoite fating from patient isolates. Front Microbiol 2022; 13:976606. [PMID: 36212849 PMCID: PMC9539820 DOI: 10.3389/fmicb.2022.976606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Plasmodium vivax, one species of parasite causing human malaria, forms a dormant liver stage, termed the hypnozoite, which activate weeks, months or years after the primary infection, causing relapse episodes. Relapses significantly contribute to the vivax malaria burden and are only killed with drugs of the 8-aminoquinoline class, which are contraindicated in many vulnerable populations. Development of new therapies targeting hypnozoites is hindered, in part, by the lack of robust methods to continuously culture and characterize this parasite. As a result, the determinants of relapse periodicity and the molecular processes that drive hypnozoite formation, persistence, and activation are largely unknown. While previous reports have described vastly different liver-stage growth metrics attributable to which hepatocyte donor lot is used to initiate culture, a comprehensive assessment of how different P. vivax patient isolates behave in the same lots at the same time is logistically challenging. Using our primary human hepatocyte-based P. vivax liver-stage culture platform, we aimed to simultaneously test the effects of how hepatocyte donor lot and P. vivax patient isolate influence the fate of sporozoites and growth of liver schizonts. We found that, while environmental factors such as hepatocyte donor lot can modulate hypnozoite formation rate, the P. vivax case is also an important determinant of the proportion of hypnozoites observed in culture. In addition, we found schizont growth to be mostly influenced by hepatocyte donor lot. These results suggest that, while host hepatocytes harbor characteristics making them more- or less-supportive of a quiescent versus growing intracellular parasite, sporozoite fating toward hypnozoites is isolate-specific. Future studies involving these host–parasite interactions, including characterization of individual P. vivax strains, should consider the impact of culture conditions on hypnozoite formation, in order to better understand this important part of the parasite’s lifecycle.
Collapse
Affiliation(s)
- Amélie Vantaux
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
- *Correspondence: Amélie Vantaux,
| | - Julie Péneau
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Caitlin A. Cooper
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
| | - Dennis E. Kyle
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
| | - Benoit Witkowski
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Steven P. Maher
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
- Steven P. Maher,
| |
Collapse
|
13
|
Pathak AK, Shiau JC, Franke-Fayard B, Shollenberger LM, Harn DA, Kyle DE, Murdock CC. Streamlining sporozoite isolation from mosquitoes by leveraging the dynamics of migration to the salivary glands. Malar J 2022; 21:264. [PMID: 36100902 PMCID: PMC9472382 DOI: 10.1186/s12936-022-04270-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 08/12/2022] [Indexed: 12/05/2022] Open
Abstract
Background Sporozoites isolated from the salivary glands of Plasmodium-infected mosquitoes are a prerequisite for several basic and pre-clinical applications. Although salivary glands are pooled to maximize sporozoite recovery, insufficient yields pose logistical and analytical hurdles; thus, predicting yields prior to isolation would be valuable. Preceding oocyst densities in the midgut is an obvious candidate. However, it is unclear whether current understanding of its relationship with sporozoite densities can be used to maximize yields, or whether it can capture the potential density-dependence in rates of sporozoite invasion of the salivary glands. Methods This study presents a retrospective analysis of Anopheles stephensi mosquitoes infected with two strains of the rodent-specific Plasmodium berghei. Mean oocyst densities were estimated in the midguts earlier in the infection (11–15 days post-blood meal), with sporozoites pooled from the salivary glands later in the infection (17–29 days). Generalized linear mixed effects models were used to determine if (1) mean oocyst densities can predict sporozoite yields from pooled salivary glands, (2) whether these densities can capture differences in rates of sporozoite invasion of salivary glands, and (3), if the interaction between oocyst densities and time could be leveraged to boost overall yields. Results The non-linear effect of mean oocyst densities confirmed the role of density-dependent constraints in limiting yields beyond certain oocyst densities. Irrespective of oocyst densities however, the continued invasion of salivary glands by the sporozoites boosted recoveries over time (17–29 days post-blood meal) for either parasite strain. Conclusions Sporozoite invasion of the salivary glands over time can be leveraged to maximize yields for P. berghei. In general, however, invasion of the salivary glands over time is a critical fitness determinant for all Plasmodium species (extrinsic incubation period, EIP). Thus, delaying sporozoite collection could, in principle, substantially reduce dissection effort for any parasite within the genus, with the results also alluding to the potential for changes in sporozoites densities over time to modify infectivity for the next host. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-022-04270-y.
Collapse
|
14
|
Valenciano AL, Gomez-Lorenzo MG, Vega-Rodríguez J, Adams JH, Roth A. In vitro models for human malaria: targeting the liver stage. Trends Parasitol 2022; 38:758-774. [PMID: 35780012 PMCID: PMC9378454 DOI: 10.1016/j.pt.2022.05.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 11/16/2022]
Abstract
The Plasmodium liver stage represents a vulnerable therapeutic target to prevent disease progression as the parasite resides in the liver before clinical representation caused by intraerythrocytic development. However, most antimalarial drugs target the blood stage of the parasite's life cycle, and the few drugs that target the liver stage are lethal to patients with a glucose-6-phosphate dehydrogenase deficiency. Furthermore, implementation of in vitro liver models to study and develop novel therapeutics against the liver stage of human Plasmodium species remains challenging. In this review, we focus on the progression of in vitro liver models developed for human Plasmodium spp. parasites, provide a brief review on important assay requirements, and lastly present recommendations to improve models to enhance the discovery process of novel preclinical therapeutics.
Collapse
Affiliation(s)
- Ana Lisa Valenciano
- Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA; Global Health Medicines R&D, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos 28760, Madrid, Spain
| | - Maria G Gomez-Lorenzo
- Global Health Medicines R&D, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos 28760, Madrid, Spain
| | - Joel Vega-Rodríguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - John H Adams
- Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA
| | - Alison Roth
- Department of Drug Discovery, Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| |
Collapse
|
15
|
Lyons FMT, Gabriela M, Tham WH, Dietrich MH. Plasmodium 6-Cysteine Proteins: Functional Diversity, Transmission-Blocking Antibodies and Structural Scaffolds. Front Cell Infect Microbiol 2022; 12:945924. [PMID: 35899047 PMCID: PMC9309271 DOI: 10.3389/fcimb.2022.945924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 06/22/2022] [Indexed: 11/30/2022] Open
Abstract
The 6-cysteine protein family is one of the most abundant surface antigens that are expressed throughout the Plasmodium falciparum life cycle. Many members of the 6-cysteine family have critical roles in parasite development across the life cycle in parasite transmission, evasion of the host immune response and host cell invasion. The common feature of the family is the 6-cysteine domain, also referred to as s48/45 domain, which is conserved across Aconoidasida. This review summarizes the current approaches for recombinant expression for 6-cysteine proteins, monoclonal antibodies against 6-cysteine proteins that block transmission and the growing collection of crystal structures that provide insights into the functional domains of this protein family.
Collapse
Affiliation(s)
- Frankie M. T. Lyons
- The Walter and Eliza Hall Institute of Medical Research, Infectious Diseases and Immune Defence Division, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Mikha Gabriela
- The Walter and Eliza Hall Institute of Medical Research, Infectious Diseases and Immune Defence Division, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Wai-Hong Tham
- The Walter and Eliza Hall Institute of Medical Research, Infectious Diseases and Immune Defence Division, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Melanie H. Dietrich
- The Walter and Eliza Hall Institute of Medical Research, Infectious Diseases and Immune Defence Division, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
16
|
Aparici Herraiz I, Caires HR, Castillo-Fernández Ó, Sima N, Méndez-Mora L, Risueño RM, Sattabongkot J, Roobsoong W, Hernández-Machado A, Fernandez-Becerra C, Barrias CC, del Portillo HA. Advancing Key Gaps in the Knowledge of Plasmodium vivax Cryptic Infections Using Humanized Mouse Models and Organs-on-Chips. Front Cell Infect Microbiol 2022; 12:920204. [PMID: 35873153 PMCID: PMC9302440 DOI: 10.3389/fcimb.2022.920204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Plasmodium vivax is the most widely distributed human malaria parasite representing 36.3% of disease burden in the South-East Asia region and the most predominant species in the region of the Americas. Recent estimates indicate that 3.3 billion of people are under risk of infection with circa 7 million clinical cases reported each year. This burden is certainly underestimated as the vast majority of chronic infections are asymptomatic. For centuries, it has been widely accepted that the only source of cryptic parasites is the liver dormant stages known as hypnozoites. However, recent evidence indicates that niches outside the liver, in particular in the spleen and the bone marrow, can represent a major source of cryptic chronic erythrocytic infections. The origin of such chronic infections is highly controversial as many key knowledge gaps remain unanswered. Yet, as parasites in these niches seem to be sheltered from immune response and antimalarial drugs, research on this area should be reinforced if elimination of malaria is to be achieved. Due to ethical and technical considerations, working with the liver, bone marrow and spleen from natural infections is very difficult. Recent advances in the development of humanized mouse models and organs-on-a-chip models, offer novel technological frontiers to study human diseases, vaccine validation and drug discovery. Here, we review current data of these frontier technologies in malaria, highlighting major challenges ahead to study P. vivax cryptic niches, which perpetuate transmission and burden.
Collapse
Affiliation(s)
- Iris Aparici Herraiz
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Hugo R. Caires
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Óscar Castillo-Fernández
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Núria Sima
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Lourdes Méndez-Mora
- Department of Condensed Matter Physics, University of Barcelona (UB), Barcelona, Spain
| | - Ruth M. Risueño
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Aurora Hernández-Machado
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
- Department of Condensed Matter Physics, University of Barcelona (UB), Barcelona, Spain
- Centre de Recerca Matemàtica (CRM), Barcelona, Spain
| | - Carmen Fernandez-Becerra
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Cristina C. Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS – Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Hernando A. del Portillo
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- *Correspondence: Hernando A. del Portillo,
| |
Collapse
|
17
|
Cova MM, Lamarque MH, Lebrun M. How Apicomplexa Parasites Secrete and Build Their Invasion Machinery. Annu Rev Microbiol 2022; 76:619-640. [PMID: 35671531 DOI: 10.1146/annurev-micro-041320-021425] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Apicomplexa are obligatory intracellular parasites that sense and actively invade host cells. Invasion is a conserved process that relies on the timely and spatially controlled exocytosis of unique specialized secretory organelles termed micronemes and rhoptries. Microneme exocytosis starts first and likely controls the intricate mechanism of rhoptry secretion. To assemble the invasion machinery, micronemal proteins-associated with the surface of the parasite-interact and form complexes with rhoptry proteins, which in turn are targeted into the host cell. This review covers the molecular advances regarding microneme and rhoptry exocytosis and focuses on how the proteins discharged from these two compartments work in synergy to drive a successful invasion event. Particular emphasis is given to the structure and molecular components of the rhoptry secretion apparatus, and to the current conceptual framework of rhoptry exocytosis that may constitute an unconventional eukaryotic secretory machinery closely related to the one described in ciliates. Expected final online publication date for the Annual Review of Microbiology, Volume 76 is September 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Marta Mendonça Cova
- Laboratory of Pathogen Host Interactions (LPHI), CNRS, University of Montpellier, Montpellier, France;
| | - Mauld H Lamarque
- Laboratory of Pathogen Host Interactions (LPHI), CNRS, University of Montpellier, Montpellier, France;
| | - Maryse Lebrun
- Laboratory of Pathogen Host Interactions (LPHI), CNRS, University of Montpellier, Montpellier, France;
| |
Collapse
|
18
|
Matteucci KC, Correa AAS, Costa DL. Recent Advances in Host-Directed Therapies for Tuberculosis and Malaria. Front Cell Infect Microbiol 2022; 12:905278. [PMID: 35669122 PMCID: PMC9163498 DOI: 10.3389/fcimb.2022.905278] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 04/21/2022] [Indexed: 11/30/2022] Open
Abstract
Tuberculosis (TB), caused by the bacterium Mycobacterium tuberculosis, and malaria, caused by parasites from the Plasmodium genus, are two of the major causes of death due to infectious diseases in the world. Both diseases are treatable with drugs that have microbicidal properties against each of the etiologic agents. However, problems related to treatment compliance by patients and emergence of drug resistant microorganisms have been a major problem for combating TB and malaria. This factor is further complicated by the absence of highly effective vaccines that can prevent the infection with either M. tuberculosis or Plasmodium. However, certain host biological processes have been found to play a role in the promotion of infection or in the pathogenesis of each disease. These processes can be targeted by host-directed therapies (HDTs), which can be administered in conjunction with the standard drug treatments for each pathogen, aiming to accelerate their elimination or to minimize detrimental side effects resulting from exacerbated inflammation. In this review we discuss potential new targets for the development of HDTs revealed by recent advances in the knowledge of host-pathogen interaction biology, and present an overview of strategies that have been tested in vivo, either in experimental models or in patients.
Collapse
Affiliation(s)
- Kely C. Matteucci
- Plataforma de Medicina Translacional Fundação Oswaldo Cruz/Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - André A. S. Correa
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Diego L. Costa
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- *Correspondence: Diego L. Costa,
| |
Collapse
|
19
|
Sánchez-Arcila JC, Jensen KDC. Forward Genetics in Apicomplexa Biology: The Host Side of the Story. Front Cell Infect Microbiol 2022; 12:878475. [PMID: 35646724 PMCID: PMC9133346 DOI: 10.3389/fcimb.2022.878475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Forward genetic approaches have been widely used in parasitology and have proven their power to reveal the complexities of host-parasite interactions in an unbiased fashion. Many aspects of the parasite's biology, including the identification of virulence factors, replication determinants, antibiotic resistance genes, and other factors required for parasitic life, have been discovered using such strategies. Forward genetic approaches have also been employed to understand host resistance mechanisms to parasitic infection. Here, we will introduce and review all forward genetic approaches that have been used to identify host factors involved with Apicomplexa infections, which include classical genetic screens and QTL mapping, GWAS, ENU mutagenesis, overexpression, RNAi and CRISPR-Cas9 library screens. Collectively, these screens have improved our understanding of host resistance mechanisms, immune regulation, vaccine and drug designs for Apicomplexa parasites. We will also discuss how recent advances in molecular genetics give present opportunities to further explore host-parasite relationships.
Collapse
Affiliation(s)
- Juan C. Sánchez-Arcila
- Department of Molecular and Cell Biology, University of California Merced, Merced, CA, United States
| | - Kirk D. C. Jensen
- Department of Molecular and Cell Biology, University of California Merced, Merced, CA, United States
- Health Science Research Institute, University of California, Merced, Merced, CA, United States
| |
Collapse
|
20
|
Mancio-Silva L, Gural N, Real E, Wadsworth MH, Butty VL, March S, Nerurkar N, Hughes TK, Roobsoong W, Fleming HE, Whittaker CA, Levine SS, Sattabongkot J, Shalek AK, Bhatia SN. A single-cell liver atlas of Plasmodium vivax infection. Cell Host Microbe 2022; 30:1048-1060.e5. [PMID: 35443155 DOI: 10.1016/j.chom.2022.03.034] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 01/31/2022] [Accepted: 03/25/2022] [Indexed: 12/15/2022]
Abstract
Malaria-causing Plasmodium vivax parasites can linger in the human liver for weeks to years and reactivate to cause recurrent blood-stage infection. Although they are an important target for malaria eradication, little is known about the molecular features of replicative and non-replicative intracellular liver-stage parasites and their host cell dependence. Here, we leverage a bioengineered human microliver platform to culture patient-derived P. vivax parasites for transcriptional profiling. Coupling enrichment strategies with bulk and single-cell analyses, we capture both parasite and host transcripts in individual hepatocytes throughout the course of infection. We define host- and state-dependent transcriptional signatures and identify unappreciated populations of replicative and non-replicative parasites that share features with sexual transmissive forms. We find that infection suppresses the transcription of key hepatocyte function genes and elicits an anti-parasite innate immune response. Our work provides a foundation for understanding host-parasite interactions and reveals insights into the biology of P. vivax dormancy and transmission.
Collapse
Affiliation(s)
- Liliana Mancio-Silva
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Institut Pasteur, Université Paris Cité, Inserm U1201, CNRS EMR9195, Unité de Biologie des Interactions Hôte-Parasite, 75015 Paris, France.
| | - Nil Gural
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Eliana Real
- Institut Pasteur, Université Paris Cité, Inserm U1201, CNRS EMR9195, Unité de Biologie des Interactions Hôte-Parasite, 75015 Paris, France
| | - Marc H Wadsworth
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; Department of Chemistry, MIT, Cambridge, MA 02139, USA
| | - Vincent L Butty
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; BioMicro Center, MIT, Cambridge, MA 02139, USA
| | - Sandra March
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Niketa Nerurkar
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Travis K Hughes
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; Department of Chemistry, MIT, Cambridge, MA 02139, USA
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine Mahidol University, Bangkok 10400, Thailand
| | - Heather E Fleming
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Charlie A Whittaker
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; BioMicro Center, MIT, Cambridge, MA 02139, USA
| | - Stuart S Levine
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; BioMicro Center, MIT, Cambridge, MA 02139, USA
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine Mahidol University, Bangkok 10400, Thailand
| | - Alex K Shalek
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Department of Chemistry, MIT, Cambridge, MA 02139, USA; Ragon Institute of Massachusetts General Hospital, MIT, and Harvard, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA.
| | - Sangeeta N Bhatia
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; The Wyss Institute for Biologically Inspired Engineering Harvard University Boston, MA 02215, USA.
| |
Collapse
|
21
|
Goswami D, Minkah NK, Kappe SHI. Malaria parasite liver stages. J Hepatol 2022; 76:735-737. [PMID: 34711453 DOI: 10.1016/j.jhep.2021.05.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/17/2021] [Accepted: 05/21/2021] [Indexed: 12/04/2022]
Affiliation(s)
- Debashree Goswami
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Nana K Minkah
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA; Department of Pediatrics, University of Washington, Seattle, WA 98105, USA
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA; Department of Pediatrics, University of Washington, Seattle, WA 98105, USA; Department of Global Health, University of Washington, Seattle, WA 98105, USA.
| |
Collapse
|
22
|
van der Boor SC, van Gemert GJ, Hanssen AEJ, van Waardenburg YM, McCall MBB, Bousema T, de Wilt JHW, Sauerwein RW, Yang ASP. Mid-Liver Stage Arrest of Plasmodium falciparum Schizonts in Primary Porcine Hepatocytes. Front Cell Infect Microbiol 2022; 12:834850. [PMID: 35252038 PMCID: PMC8892583 DOI: 10.3389/fcimb.2022.834850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
During co-evolution Plasmodium parasites and vertebrates went through a process of selection resulting in defined and preferred parasite-host combinations. As such, Plasmodium falciparum (Pf) sporozoites can infect human hepatocytes while seemingly incompatible with host cellular machinery of other species. The compatibility between parasite invasion ligands and their respective human hepatocyte receptors plays a key role in Pf host selectivity. However, it is unclear whether the ability of Pf sporozoites to mature in cross-species infection also plays a role in host tropism. Here we used fresh hepatocytes isolated from porcine livers to study permissiveness to Pf sporozoite invasion and development. We monitored intra-hepatic development via immunofluorescence using anti-HSP70, MSP1, EXP1, and EXP2 antibodies. Our data shows that Pf sporozoites can invade non-human hepatocytes and undergo partial maturation with a significant decrease in schizont numbers between day three and day five. A possible explanation is that Pf sporozoites fail to form a parasitophorous vacuolar membrane (PVM) during invasion. Indeed, the observed aberrant EXP1 and EXP2 staining supports the presence of an atypical PVM. Functions of the PVM include the transport of nutrients, export of waste, and offering a protective barrier against intracellular host effectors. Therefore, an atypical PVM likely results in deficiencies that may detrimentally impact parasite development at multiple levels. In summary, despite successful invasion of porcine hepatocytes, Pf development arrests at mid-stage, possibly due to an inability to mobilize critical nutrients across the PVM. These findings underscore the potential of a porcine liver model for understanding the importance of host factors required for Pf mid-liver stage development.
Collapse
Affiliation(s)
- Saskia C. van der Boor
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Geert-Jan van Gemert
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Alex E. J. Hanssen
- Animal Research Facility, Radboud University Medical Center, Nijmegen, Netherlands
| | - Youri M. van Waardenburg
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Matthew B. B. McCall
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Teun Bousema
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Robert W. Sauerwein
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
- TropIQ Health Sciences, Nijmegen, Netherlands
| | - Annie S. P. Yang
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
23
|
Dietrich MH, Chan LJ, Adair A, Boulet C, O'Neill MT, Tan LL, Keremane S, Mok YF, Lo AW, Gilson P, Tham WH. Structure of the Pf12 and Pf41 heterodimeric complex of Plasmodium falciparum 6-cysteine proteins. FEMS MICROBES 2022; 3:xtac005. [PMID: 35308105 PMCID: PMC8930183 DOI: 10.1093/femsmc/xtac005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 01/19/2022] [Accepted: 02/10/2022] [Indexed: 11/16/2022] Open
Abstract
During the different stages of the Plasmodium life cycle, surface-associated proteins establish key interactions with the host and play critical roles in parasite survival. The 6-cysteine (6-cys) protein family is one of the most abundant surface antigens and expressed throughout the Plasmodium falciparum life cycle. This protein family is conserved across Plasmodium species and plays critical roles in parasite transmission, evasion of the host immune response and host cell invasion. Several 6-cys proteins are present on the parasite surface as hetero-complexes but it is not known how two 6-cys proteins interact together. Here, we present a crystal structure of Pf12 bound to Pf41 at 2.85 Å resolution, two P. falciparum proteins usually found on the parasite surface of late schizonts and merozoites. Our structure revealed two critical interfaces required for complex formation with important implications on how different 6-cysteine proteins may interact with each other. Using structure-function analyses, we identified important residues for Pf12-Pf41 complex formation. In addition, we generated 16 nanobodies against Pf12 and Pf41 and showed that several Pf12-specific nanobodies inhibit Pf12-Pf41 complex formation. Using X-ray crystallography, we were able to describe the structural mechanism of an inhibitory nanobody in blocking Pf12-Pf41 complex formation. Future studies using these inhibitory nanobodies will be useful to determine the functional role of these two 6-cys proteins in malaria parasites.
Collapse
Affiliation(s)
- Melanie H Dietrich
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Li-Jin Chan
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Amy Adair
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | | | - Matthew T O'Neill
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Li Lynn Tan
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Sravya Keremane
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Yee-Foong Mok
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, Melbourne, Australia
| | - Alvin W Lo
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | | | - Wai-Hong Tham
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
24
|
Cha SJ, Kim MS, Na CH, Jacobs-Lorena M. Plasmodium sporozoite phospholipid scramblase interacts with mammalian carbamoyl-phosphate synthetase 1 to infect hepatocytes. Nat Commun 2021; 12:6773. [PMID: 34799567 PMCID: PMC8604956 DOI: 10.1038/s41467-021-27109-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 11/04/2021] [Indexed: 11/14/2022] Open
Abstract
After inoculation by the bite of an infected mosquito, Plasmodium sporozoites enter the blood stream and infect the liver, where each infected cell produces thousands of merozoites. These in turn, infect red blood cells and cause malaria symptoms. To initiate a productive infection, sporozoites must exit the circulation by traversing the blood lining of the liver vessels after which they infect hepatocytes with unique specificity. We screened a phage display library for peptides that structurally mimic (mimotope) a sporozoite ligand for hepatocyte recognition. We identified HP1 (hepatocyte-binding peptide 1) that mimics a ~50 kDa sporozoite ligand (identified as phospholipid scramblase). Further, we show that HP1 interacts with a ~160 kDa hepatocyte membrane putative receptor (identified as carbamoyl-phosphate synthetase 1). Importantly, immunization of mice with the HP1 peptide partially protects them from infection by the rodent parasite P. berghei. Moreover, an antibody to the HP1 mimotope inhibits human parasite P. falciparum infection of human hepatocytes in culture. The sporozoite ligand for hepatocyte invasion is a potential novel pre-erythrocytic vaccine candidate. After transmission of Plasmodium sporozoites from infected mosquitoes, parasites first infect hepatocytes. Here, Cha et al. identify a sporozoite ligand (phospholipid scramblase) and the hepatocytic receptor (carbamoyl-phosphate synthetase 1) as relevant for hepatocyte invasion and show that an antibody to hepatocyte-binding peptide 1 (HP1), which structurally mimics the sporozoite ligand, partially protects mice from infection.
Collapse
Affiliation(s)
- Sung-Jae Cha
- Johns Hopkins Bloomberg School of Public Health, Department of Molecular Microbiology and Immunology and Malaria Research Institute, 615N. Wolfe St., Baltimore, MD, 21205, USA.
| | - Min-Sik Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Chan Hyun Na
- Department of Neurology, Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Marcelo Jacobs-Lorena
- Johns Hopkins Bloomberg School of Public Health, Department of Molecular Microbiology and Immunology and Malaria Research Institute, 615N. Wolfe St., Baltimore, MD, 21205, USA.
| |
Collapse
|
25
|
Abstract
Host-directed therapy (HDT) is gaining traction as a strategy to combat infectious diseases caused by viruses and intracellular bacteria, but its implementation in the context of parasitic diseases has received less attention. Here, we provide a brief overview of this field and advocate HDT as a promising strategy for antimalarial intervention based on untapped targets. HDT provides a basis from which repurposed drugs could be rapidly deployed and is likely to strongly limit the emergence of resistance. This strategy can be applied to any intracellular pathogen and is particularly well placed in situations in which rapid identification of treatments is needed, such as emerging infections and pandemics, as starkly illustrated by the current COVID-19 crisis.
Collapse
|
26
|
Kolli SK, Salman AM, Ramesar J, Chevalley-Maurel S, Kroeze H, Geurten FGA, Miyazaki S, Mukhopadhyay E, Marin-Mogollon C, Franke-Fayard B, Hill AVS, Janse CJ. Screening of viral-vectored P. falciparum pre-erythrocytic candidate vaccine antigens using chimeric rodent parasites. PLoS One 2021; 16:e0254498. [PMID: 34252120 PMCID: PMC8274855 DOI: 10.1371/journal.pone.0254498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/28/2021] [Indexed: 11/19/2022] Open
Abstract
To screen for additional vaccine candidate antigens of Plasmodium pre-erythrocytic stages, fourteen P. falciparum proteins were selected based on expression in sporozoites or their role in establishment of hepatocyte infection. For preclinical evaluation of immunogenicity of these proteins in mice, chimeric P. berghei sporozoites were created that express the P. falciparum proteins in sporozoites as an additional copy gene under control of the uis4 gene promoter. All fourteen chimeric parasites produced sporozoites but sporozoites of eight lines failed to establish a liver infection, indicating a negative impact of these P. falciparum proteins on sporozoite infectivity. Immunogenicity of the other six proteins (SPELD, ETRAMP10.3, SIAP2, SPATR, HT, RPL3) was analyzed by immunization of inbred BALB/c and outbred CD-1 mice with viral-vectored (ChAd63 or ChAdOx1, MVA) vaccines, followed by challenge with chimeric sporozoites. Protective immunogenicity was determined by analyzing parasite liver load and prepatent period of blood stage infection after challenge. Of the six proteins only SPELD immunized mice showed partial protection. We discuss both the low protective immunogenicity of these proteins in the chimeric rodent malaria challenge model and the negative effect on P. berghei sporozoite infectivity of several P. falciparum proteins expressed in the chimeric sporozoites.
Collapse
Affiliation(s)
- Surendra Kumar Kolli
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Ahmed M. Salman
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Jai Ramesar
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Hans Kroeze
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Fiona G. A. Geurten
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Shinya Miyazaki
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Ekta Mukhopadhyay
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | | - Adrian V. S. Hill
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Chris J. Janse
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
27
|
Vijayan K, Wei L, Glennon EKK, Mattocks C, Bourgeois N, Staker B, Kaushansky A. Host-targeted Interventions as an Exciting Opportunity to Combat Malaria. Chem Rev 2021; 121:10452-10468. [PMID: 34197083 DOI: 10.1021/acs.chemrev.1c00062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Terminal and benign diseases alike in adults, children, pregnant women, and others are successfully treated by pharmacological inhibitors that target human enzymes. Despite extensive global efforts to fight malaria, the disease continues to be a massive worldwide health burden, and new interventional strategies are needed. Current drugs and vector control strategies have contributed to the reduction in malaria deaths over the past 10 years, but progress toward eradication has waned in recent years. Resistance to antimalarial drugs is a substantial and growing problem. Moreover, targeting dormant forms of the malaria parasite Plasmodium vivax is only possible with two approved drugs, which are both contraindicated for individuals with glucose-6-phosphate dehydrogenase deficiency and in pregnant women. Plasmodium parasites are obligate intracellular parasites and thus have specific and absolute requirements of their hosts. Growing evidence has described these host necessities, paving the way for opportunities to pharmacologically target host factors to eliminate Plasmodium infection. Here, we describe progress in malaria research and adjacent fields and discuss key challenges that remain in implementing host-directed therapy against malaria.
Collapse
Affiliation(s)
| | - Ling Wei
- Seattle Children's Research Institute, Seattle, Washington 98109, United States
| | | | - Christa Mattocks
- Department of Global Health, University of Washington, Seattle, Washington 98195, United States
| | - Natasha Bourgeois
- Seattle Children's Research Institute, Seattle, Washington 98109, United States.,Department of Global Health, University of Washington, Seattle, Washington 98195, United States
| | - Bart Staker
- Seattle Children's Research Institute, Seattle, Washington 98109, United States
| | - Alexis Kaushansky
- Seattle Children's Research Institute, Seattle, Washington 98109, United States.,Department of Global Health, University of Washington, Seattle, Washington 98195, United States.,Department of Pediatrics, University of Washington, Seattle, Washington 98105, United States.,Brotman Baty Institute for Precision Medicine, Seattle, Washington 98195, United States
| |
Collapse
|
28
|
Amanzougaghene N, Tajeri S, Yalaoui S, Lorthiois A, Soulard V, Gego A, Rametti A, Risco-Castillo V, Moreno A, Tefit M, van Gemert GJ, Sauerwein RW, Vaillant JC, Ravassard P, Pérignon JL, Froissard P, Mazier D, Franetich JF. The Host Protein Aquaporin-9 is Required for Efficient Plasmodium falciparum Sporozoite Entry into Human Hepatocytes. Front Cell Infect Microbiol 2021; 11:704662. [PMID: 34268141 PMCID: PMC8276244 DOI: 10.3389/fcimb.2021.704662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/02/2021] [Indexed: 12/11/2022] Open
Abstract
Hepatocyte invasion by Plasmodium sporozoites represents a promising target for innovative antimalarial therapy, but the molecular events mediating this process are still largely uncharacterized. We previously showed that Plasmodium falciparum sporozoite entry into hepatocytes strictly requires CD81. However, CD81-overexpressing human hepatoma cells remain refractory to P. falciparum infection, suggesting the existence of additional host factors necessary for sporozoite entry. Here, through differential transcriptomic analysis of human hepatocytes and hepatoma HepG2-CD81 cells, the transmembrane protein Aquaporin-9 (AQP9) was found to be among the most downregulated genes in hepatoma cells. RNA silencing showed that sporozoite invasion of hepatocytes requires AQP9 expression. AQP9 overexpression in hepatocytes increased their permissiveness to P. falciparum. Moreover, chemical disruption with the AQP9 inhibitor phloretin markedly inhibited hepatocyte infection. Our findings identify AQP9 as a novel host factor required for P. falciparum sporozoite hepatocyte-entry and indicate that AQP9 could be a potential therapeutic target.
Collapse
Affiliation(s)
- Nadia Amanzougaghene
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Shahin Tajeri
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Samir Yalaoui
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | - Audrey Lorthiois
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | - Valérie Soulard
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Audrey Gego
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | - Armelle Rametti
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | | | - Alicia Moreno
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Maurel Tefit
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, MMB-NCMLS, Nijmegen, Netherlands
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, MMB-NCMLS, Nijmegen, Netherlands
| | - Jean-Christophe Vaillant
- AP-HP, Service de Chirurgie Digestive, Hépato-Bilio-Pancréatique et Transplantation Hépatique, Centre Hospitalo-Universitaire Pitié-Salpêtrière, Paris, France
| | - Philippe Ravassard
- CR-ICM - LGN CNRS UMR-7991, IFR des Neurosciences, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Jean-Louis Pérignon
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | - Patrick Froissard
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | - Dominique Mazier
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Jean-François Franetich
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| |
Collapse
|
29
|
Nanobody generation and structural characterization of Plasmodium falciparum 6-cysteine protein Pf12p. Biochem J 2021; 478:579-595. [PMID: 33480416 PMCID: PMC7886318 DOI: 10.1042/bcj20200415] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 01/09/2023]
Abstract
Surface-associated proteins play critical roles in the Plasmodium parasite life cycle and are major targets for vaccine development. The 6-cysteine (6-cys) protein family is expressed in a stage-specific manner throughout Plasmodium falciparum life cycle and characterized by the presence of 6-cys domains, which are β-sandwich domains with conserved sets of disulfide bonds. Although several 6-cys family members have been implicated to play a role in sexual stages, mosquito transmission, evasion of the host immune response and host cell invasion, the precise function of many family members is still unknown and structural information is only available for four 6-cys proteins. Here, we present to the best of our knowledge, the first crystal structure of the 6-cys protein Pf12p determined at 2.8 Å resolution. The monomeric molecule folds into two domains, D1 and D2, both of which adopt the canonical 6-cys domain fold. Although the structural fold is similar to that of Pf12, its paralog in P. falciparum, we show that Pf12p does not complex with Pf41, which is a known interaction partner of Pf12. We generated 10 distinct Pf12p-specific nanobodies which map into two separate epitope groups; one group which binds within the D2 domain, while several members of the second group bind at the interface of the D1 and D2 domain of Pf12p. Characterization of the structural features of the 6-cys family and their associated nanobodies provide a framework for generating new tools to study the diverse functions of the 6-cys protein family in the Plasmodium life cycle.
Collapse
|
30
|
Steel RWJ, Vigdorovich V, Dambrauskas N, Wilder BK, Arredondo SA, Goswami D, Kumar S, Carbonetti S, Swearingen KE, Nguyen T, Betz W, Camargo N, Fisher BS, Soden J, Thomas H, Freeth J, Moritz RL, Noah Sather D, Kappe SHI. Platelet derived growth factor receptor β (PDGFRβ) is a host receptor for the human malaria parasite adhesin TRAP. Sci Rep 2021; 11:11328. [PMID: 34059712 PMCID: PMC8166973 DOI: 10.1038/s41598-021-90722-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/13/2021] [Indexed: 02/04/2023] Open
Abstract
Following their inoculation by the bite of an infected Anopheles mosquito, the malaria parasite sporozoite forms travel from the bite site in the skin into the bloodstream, which transports them to the liver. The thrombospondin-related anonymous protein (TRAP) is a type 1 transmembrane protein that is released from secretory organelles and relocalized on the sporozoite plasma membrane. TRAP is required for sporozoite motility and host infection, and its extracellular portion contains adhesive domains that are predicted to engage host receptors. Here, we identified the human platelet-derived growth factor receptor β (hPDGFRβ) as one such protein receptor. Deletion constructs showed that the von Willebrand factor type A and thrombospondin repeat domains of TRAP are both required for optimal binding to hPDGFRβ-expressing cells. We also demonstrate that this interaction is conserved in the human-infective parasite Plasmodium vivax, but not the rodent-infective parasite Plasmodium yoelii. We observed expression of hPDGFRβ mainly in cells associated with the vasculature suggesting that TRAP:hPDGFRβ interaction may play a role in the recognition of blood vessels by invading sporozoites.
Collapse
Affiliation(s)
- Ryan W J Steel
- Seattle Children's Research Institute, Seattle, WA, USA
- Infectious Diseases and Immune Defence Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | | | | | - Brandon K Wilder
- Seattle Children's Research Institute, Seattle, WA, USA
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, 97006, USA
| | | | | | - Sudhir Kumar
- Seattle Children's Research Institute, Seattle, WA, USA
| | | | | | - Thao Nguyen
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Will Betz
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Nelly Camargo
- Seattle Children's Research Institute, Seattle, WA, USA
| | | | - Jo Soden
- Retrogenix Ltd, Chinley, High Peak, SK23 6FJ, UK
| | - Helen Thomas
- Retrogenix Ltd, Chinley, High Peak, SK23 6FJ, UK
| | - Jim Freeth
- Retrogenix Ltd, Chinley, High Peak, SK23 6FJ, UK
| | | | - D Noah Sather
- Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| | - Stefan H I Kappe
- Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| |
Collapse
|
31
|
Kulkeaw K. Next-Generation Human Liver Models for Antimalarial Drug Assays. Antibiotics (Basel) 2021; 10:antibiotics10060642. [PMID: 34071885 PMCID: PMC8229011 DOI: 10.3390/antibiotics10060642] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 12/23/2022] Open
Abstract
Advances in malaria prevention and treatment have significantly reduced the related morbidity and mortality worldwide, however, malaria continues to be a major threat to global public health. Because Plasmodium parasites reside in the liver prior to the appearance of clinical manifestations caused by intraerythrocytic development, the Plasmodium liver stage represents a vulnerable therapeutic target to prevent progression. Currently, a small number of drugs targeting liver-stage parasites are available, but all cause lethal side effects in glucose-6-phosphate dehydrogenase-deficient individuals, emphasizing the necessity for new drug development. Nevertheless, a longstanding hurdle to developing new drugs is the availability of appropriate in vitro cultures, the crucial conventional platform for evaluating the efficacy and toxicity of drugs in the preclinical phase. Most current cell culture systems rely primarily on growing immortalized or cancerous cells in the form of a two-dimensional monolayer, which is not very physiologically relevant to the complex cellular architecture of the human body. Although primary human cells are more relevant to human physiology, they are mainly hindered by batch-to-batch variation, limited supplies, and ethical issues. Advances in stem cell technologies and multidimensional culture have allowed the modelling of human infectious diseases. Here, current in vitro hepatic models and toolboxes for assaying the antimalarial drug activity are summarized. Given the physiological potential of pluripotent and adult stem cells to model liver-stage malaria, the opportunities and challenges in drug development against liver-stage malaria is highlighted, paving the way to assess the efficacy of hepatic plasmodicidal activity.
Collapse
Affiliation(s)
- Kasem Kulkeaw
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
32
|
Loubens M, Vincensini L, Fernandes P, Briquet S, Marinach C, Silvie O. Plasmodium sporozoites on the move: Switching from cell traversal to productive invasion of hepatocytes. Mol Microbiol 2021; 115:870-881. [PMID: 33191548 PMCID: PMC8247013 DOI: 10.1111/mmi.14645] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/18/2022]
Abstract
Parasites of the genus Plasmodium, the etiological agent of malaria, are transmitted through the bite of anopheline mosquitoes, which deposit sporozoites into the host skin. Sporozoites migrate through the dermis, enter the bloodstream, and rapidly traffic to the liver. They cross the liver sinusoidal barrier and traverse several hepatocytes before switching to productive invasion of a final one for replication inside a parasitophorous vacuole. Cell traversal and productive invasion are functionally independent processes that require proteins secreted from specialized secretory organelles known as micronemes. In this review, we summarize the current understanding of how sporozoites traverse through cells and productively invade hepatocytes, and discuss the role of environmental sensing in switching from a migratory to an invasive state. We propose that timely controlled secretion of distinct microneme subsets could play a key role in successful migration and infection of hepatocytes. A better understanding of these essential biological features of the Plasmodium sporozoite may contribute to the development of new strategies to fight against the very first and asymptomatic stage of malaria.
Collapse
Affiliation(s)
- Manon Loubens
- Centre d’Immunologie et des Maladies InfectieusesSorbonne Université, INSERM, CNRS, CIMI‐ParisParisFrance
| | - Laetitia Vincensini
- Centre d’Immunologie et des Maladies InfectieusesSorbonne Université, INSERM, CNRS, CIMI‐ParisParisFrance
| | - Priyanka Fernandes
- Centre d’Immunologie et des Maladies InfectieusesSorbonne Université, INSERM, CNRS, CIMI‐ParisParisFrance
| | - Sylvie Briquet
- Centre d’Immunologie et des Maladies InfectieusesSorbonne Université, INSERM, CNRS, CIMI‐ParisParisFrance
| | - Carine Marinach
- Centre d’Immunologie et des Maladies InfectieusesSorbonne Université, INSERM, CNRS, CIMI‐ParisParisFrance
| | - Olivier Silvie
- Centre d’Immunologie et des Maladies InfectieusesSorbonne Université, INSERM, CNRS, CIMI‐ParisParisFrance
| |
Collapse
|
33
|
Livingstone MC, Bitzer AA, Giri A, Luo K, Sankhala RS, Choe M, Zou X, Dennison SM, Li Y, Washington W, Ngauy V, Tomaras GD, Joyce MG, Batchelor AH, Dutta S. In vitro and in vivo inhibition of malaria parasite infection by monoclonal antibodies against Plasmodium falciparum circumsporozoite protein (CSP). Sci Rep 2021; 11:5318. [PMID: 33674699 PMCID: PMC7970865 DOI: 10.1038/s41598-021-84622-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 02/11/2021] [Indexed: 01/23/2023] Open
Abstract
Plasmodium falciparum malaria contributes to a significant global disease burden. Circumsporozoite protein (CSP), the most abundant sporozoite stage antigen, is a prime vaccine candidate. Inhibitory monoclonal antibodies (mAbs) against CSP map to either a short junctional sequence or the central (NPNA)n repeat region. We compared in vitro and in vivo activities of six CSP-specific mAbs derived from human recipients of a recombinant CSP vaccine RTS,S/AS01 (mAbs 317 and 311); an irradiated whole sporozoite vaccine PfSPZ (mAbs CIS43 and MGG4); or individuals exposed to malaria (mAbs 580 and 663). RTS,S mAb 317 that specifically binds the (NPNA)n epitope, had the highest affinity and it elicited the best sterile protection in mice. The most potent inhibitor of sporozoite invasion in vitro was mAb CIS43 which shows dual-specific binding to the junctional sequence and (NPNA)n. In vivo mouse protection was associated with the mAb reactivity to the NANPx6 peptide, the in vitro inhibition of sporozoite invasion activity, and kinetic parameters measured using intact mAbs or their Fab fragments. Buried surface area between mAb and its target epitope was also associated with in vivo protection. Association and disconnects between in vitro and in vivo readouts has important implications for the design and down-selection of the next generation of CSP based interventions.
Collapse
Affiliation(s)
- Merricka C Livingstone
- Structural Vaccinology Lab, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Alexis A Bitzer
- Structural Vaccinology Lab, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Alish Giri
- Structural Vaccinology Lab, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Kun Luo
- Structural Vaccinology Lab, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Rajeshwer S Sankhala
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Misook Choe
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Xiaoyan Zou
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, USA
| | - S Moses Dennison
- Center for Human Systems Immunology, Duke University Medical Center, Durham, NC, USA
- Departments of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Yuanzhang Li
- Statistics and Epidemiology Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - William Washington
- Statistics and Epidemiology Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Viseth Ngauy
- Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Georgia D Tomaras
- Center for Human Systems Immunology, Duke University Medical Center, Durham, NC, USA
- Departments of Surgery, Duke University Medical Center, Durham, NC, USA
- Departments of Immunology, Duke University Medical Center, Durham, NC, USA
- Departments of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - M Gordon Joyce
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Adrian H Batchelor
- Structural Vaccinology Lab, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Sheetij Dutta
- Structural Vaccinology Lab, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA.
| |
Collapse
|
34
|
Hamada S, Pionneau C, Parizot C, Silvie O, Chardonnet S, Marinach C. In-depth proteomic analysis of Plasmodium berghei sporozoites using trapped ion mobility spectrometry with parallel accumulation-serial fragmentation. Proteomics 2021; 21:e2000305. [PMID: 33452840 DOI: 10.1002/pmic.202000305] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 11/11/2022]
Abstract
Sporozoites of the malaria parasite Plasmodium are transmitted by mosquitoes and infect the liver for an initial and obligatory round of replication, before exponential multiplication in the blood and onset of the disease. Sporozoites and liver stages provide attractive targets for malaria vaccines and prophylactic drugs. In this context, defining the parasite proteome is important to explore the parasite biology and to identify potential targets for antimalarial strategies. Previous studies have determined the total proteome of sporozoites from the two main human malaria parasites, P. falciparum and P. vivax, as well as P. yoelii, which infects rodents. Another murine malaria parasite, P. berghei, is widely used to investigate the parasite biology. However, a deep view of the proteome of P. berghei sporozoites is still missing. To fill this gap, we took advantage of the highly sensitive timsTOF PRO mass spectrometer, combined with three alternative methods for sporozoite purification, to identify the proteome of P. berghei sporozoites using low numbers of parasites. This study provides a reference proteome for P. berghei sporozoites, identifying a core set of proteins expressed across species, and illustrates how the unprecedented sensitivity of the timsTOF PRO system enables deep proteomic analysis from limited sample amounts.
Collapse
Affiliation(s)
- Soumia Hamada
- Sorbonne Université, INSERM, UMS PASS, Plateforme Post-génomique de la Pitié Salpêtrière (P3S), Paris, France.,Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Cédric Pionneau
- Sorbonne Université, INSERM, UMS PASS, Plateforme Post-génomique de la Pitié Salpêtrière (P3S), Paris, France
| | - Christophe Parizot
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Assistance Publique Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Olivier Silvie
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Solenne Chardonnet
- Sorbonne Université, INSERM, UMS PASS, Plateforme Post-génomique de la Pitié Salpêtrière (P3S), Paris, France
| | - Carine Marinach
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| |
Collapse
|
35
|
Arredondo SA, Schepis A, Reynolds L, Kappe SHI. Secretory Organelle Function in the Plasmodium Sporozoite. Trends Parasitol 2021; 37:651-663. [PMID: 33589364 DOI: 10.1016/j.pt.2021.01.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/20/2021] [Accepted: 01/20/2021] [Indexed: 12/14/2022]
Abstract
Plasmodium sporozoites exhibit a complex infection biology in the mosquito and mammalian hosts. The sporozoite apical secretory organelles, the micronemes and rhoptries, store protein mediators of parasite/host/vector interactions and must secrete them in a temporally and spatially well orchestrated manner. Micronemal proteins are critical for sporozoite motility throughout its journey from the mosquito midgut oocyst to the mammalian liver, and also for cell traversal (CT) and hepatocyte invasion. Rhoptry proteins, until recently thought to be only important for hepatocyte invasion, appear to also play an unexpected role in motility and in the interaction with mosquito tissue. Therefore, navigating the different microenvironments with secretion likely requires the sporozoite to have a more complex system of secretory organelles than previously appreciated.
Collapse
Affiliation(s)
- Silvia A Arredondo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Antonino Schepis
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Laura Reynolds
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA; Department of Pediatrics, University of Washington, Seattle, WA 98105, USA; Department of Global Health, University of Washington, Seattle, WA 98105, USA.
| |
Collapse
|
36
|
Knöckel J, Dundas K, Yang ASP, Galaway F, Metcalf T, Gemert GJV, Sauerwein RW, Rayner JC, Billker O, Wright GJ. Systematic Identification of Plasmodium Falciparum Sporozoite Membrane Protein Interactions Reveals an Essential Role for the p24 Complex in Host Infection. Mol Cell Proteomics 2021; 20:100038. [PMID: 33515807 PMCID: PMC7950211 DOI: 10.1074/mcp.ra120.002432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 12/22/2020] [Indexed: 12/14/2022] Open
Abstract
Sporozoites are a motile form of malaria-causing Plasmodium falciparum parasites that migrate from the site of transmission in the dermis through the bloodstream to invade hepatocytes. Sporozoites interact with many cells within the host, but the molecular identity of these interactions and their role in the pathology of malaria is poorly understood. Parasite proteins that are secreted and embedded within membranes are known to be important for these interactions, but our understanding of how they interact with each other to form functional complexes is largely unknown. Here, we compile a library of recombinant proteins representing the repertoire of cell surface and secreted proteins from the P. falciparum sporozoite and use an assay designed to detect extracellular interactions to systematically identify complexes. We identify three protein complexes including an interaction between two components of the p24 complex that is involved in the trafficking of glycosylphosphatidylinositol-anchored proteins through the secretory pathway. Plasmodium parasites lacking either gene are strongly inhibited in the establishment of liver-stage infections. These findings reveal an important role for the p24 complex in malaria pathogenesis and show that the library of recombinant proteins represents a valuable resource to investigate P. falciparum sporozoite biology.
Collapse
Key Words
- avexis, avidity-based extracellular interaction screen
- csp, circumsporozoite protein
- gpi, glycosylphosphatidylinositol
- hbs, hepes-buffered saline
- hek, human embryonic kidney
- ivis, in vivo imaging system
- msp, merozoite surface protein
- piesp15, parasite-infected erythrocyte surface protein 15
- spr, surface plasmon resonance
- trap, thrombospondin-related anonymous protein
Collapse
Affiliation(s)
- Julia Knöckel
- Cell Surface Signalling Laboratory, Wellcome Sanger Institute, Cambridge, United Kingdom; Malaria Programme, Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Kirsten Dundas
- Cell Surface Signalling Laboratory, Wellcome Sanger Institute, Cambridge, United Kingdom; Malaria Programme, Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Annie S P Yang
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Francis Galaway
- Cell Surface Signalling Laboratory, Wellcome Sanger Institute, Cambridge, United Kingdom; Malaria Programme, Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Tom Metcalf
- Malaria Programme, Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Geert-Jan van Gemert
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Robert W Sauerwein
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Julian C Rayner
- Malaria Programme, Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Oliver Billker
- Malaria Programme, Wellcome Sanger Institute, Cambridge, United Kingdom; The Laboratory for Molecular Infection Medicine Sweden (MIMS) and Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Gavin J Wright
- Cell Surface Signalling Laboratory, Wellcome Sanger Institute, Cambridge, United Kingdom; Malaria Programme, Wellcome Sanger Institute, Cambridge, United Kingdom; Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York, York, United Kingdom.
| |
Collapse
|
37
|
Mello-Vieira J, Enguita FJ, de Koning-Ward TF, Zuzarte-Luís V, Mota MM. Plasmodium translocon component EXP2 facilitates hepatocyte invasion. Nat Commun 2020; 11:5654. [PMID: 33159090 PMCID: PMC7648069 DOI: 10.1038/s41467-020-19492-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022] Open
Abstract
Plasmodium parasites possess a translocon that exports parasite proteins into the infected erythrocyte. Although the translocon components are also expressed during the mosquito and liver stage of infection, their function remains unexplored. Here, using a combination of genetic and chemical assays, we show that the translocon component Exported Protein 2 (EXP2) is critical for invasion of hepatocytes. EXP2 is a pore-forming protein that is secreted from the sporozoite upon contact with the host cell milieu. EXP2-deficient sporozoites are impaired in invasion, which can be rescued by the exogenous administration of recombinant EXP2 and alpha-hemolysin (an S. aureus pore-forming protein), as well as by acid sphingomyelinase. The latter, together with the negative impact of chemical and genetic inhibition of acid sphingomyelinase on invasion, reveals that EXP2 pore-forming activity induces hepatocyte membrane repair, which plays a key role in parasite invasion. Overall, our findings establish a novel and critical function for EXP2 that leads to an active participation of the host cell in Plasmodium sporozoite invasion, challenging the current view of the establishment of liver stage infection. While the role of Plasmodium EXP2 protein as translocon component of blood stage parasites is established, its functional role in liver stage parasites remains unclear. Here, Mello-Vieira et al. reveal that EXP2 pore-forming activity induces hepatocyte membrane repair and hence is critical for hepatocyte invasion.
Collapse
Affiliation(s)
- João Mello-Vieira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Francisco J Enguita
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | | | - Vanessa Zuzarte-Luís
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028, Lisboa, Portugal.
| | - Maria M Mota
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028, Lisboa, Portugal.
| |
Collapse
|
38
|
Karam J, Méresse S, Kremer L, Daher W. The roles of tetraspanins in bacterial infections. Cell Microbiol 2020; 22:e13260. [PMID: 32902857 DOI: 10.1111/cmi.13260] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/22/2020] [Accepted: 07/24/2020] [Indexed: 12/18/2022]
Abstract
Tetraspanins, a wide family composed of 33 transmembrane proteins, are associated with different types of proteins through which they arbitrate important cellular processes such as fusion, adhesion, invasion, tissue differentiation and immunological responses. Tetraspanins share a comparable structural design, which consists of four hydrophobic transmembrane domains with cytoplasmic and extracellular loops. They cooperate with different proteins, including other tetraspanins, receptors or signalling proteins to compose functional complexes at the cell surface, designated tetraspanin-enriched microdomains (TEM). Increasing evidences establish that tetraspanins are exploited by numerous intracellular pathogens as a doorway for entering and replicating within human cells. Although previous surveys focused mainly on viruses and parasites, it is now becoming clear that bacteria interact with tetraspanins, using TEM as a "gateway" to infection. In this review, we examine the biological functions of tetraspanins that are relevant to bacterial infective procedures and consider the available data that reveal how different bacteria benefit from host cell tetraspanins in infection and in the pathogenesis of diseases. We will also emphasise the stimulating potentials of targeting tetraspanins for preventing bacterial infectious diseases, using specific neutralising antibodies or anti-adhesion peptide-based therapies. Such innovative therapeutic opportunities may deliver alternatives for fighting difficult-to-manage and drug-resistant bacterial pathogens.
Collapse
Affiliation(s)
- Jona Karam
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France
| | | | - Laurent Kremer
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France.,INSERM, IRIM, Montpellier, France
| | - Wassim Daher
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France.,INSERM, IRIM, Montpellier, France
| |
Collapse
|
39
|
Subramani PA, Vartak-Sharma N, Sreekumar S, Mathur P, Nayer B, Dakhore S, Basavanna SK, Kalappa DM, Krishnamurthy RV, Mukhi B, Mishra P, Yoshida N, Ghosh SK, Shandil R, Narayanan S, Campo B, Hasegawa K, Anvikar AR, Valecha N, Sundaramurthy V. Plasmodium vivax liver stage assay platforms using Indian clinical isolates. Malar J 2020; 19:214. [PMID: 32571333 PMCID: PMC7310233 DOI: 10.1186/s12936-020-03284-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/12/2020] [Indexed: 12/24/2022] Open
Abstract
Background Vivax malaria is associated with significant morbidity and economic loss, and constitutes the bulk of malaria cases in large parts of Asia and South America as well as recent case reports in Africa. The widespread prevalence of vivax is a challenge to global malaria elimination programmes. Vivax malaria control is particularly challenged by existence of dormant liver stage forms that are difficult to treat and are responsible for multiple relapses, growing drug resistance to the asexual blood stages and host-genetic factors that preclude use of specific drugs like primaquine capable of targeting Plasmodium vivax liver stages. Despite an obligatory liver-stage in the Plasmodium life cycle, both the difficulty in obtaining P. vivax sporozoites and the limited availability of robust host cell models permissive to P. vivax infection are responsible for the limited knowledge of hypnozoite formation biology and relapse mechanisms, as well as the limited capability to do drug screening. Although India accounts for about half of vivax malaria cases world-wide, very little is known about the vivax liver stage forms in the context of Indian clinical isolates. Methods To address this, methods were established to obtain infective P. vivax sporozoites from an endemic region in India and multiple assay platforms set up to detect and characterize vivax liver stage forms. Different hepatoma cell lines, including the widely used HCO4 cells, primary human hepatocytes as well as hepatocytes obtained from iPSC’s generated from vivax patients and healthy donors were tested for infectivity with P. vivax sporozoites. Results Both large and small forms of vivax liver stage are detected in these assays, although the infectivity obtained in these platforms are low. Conclusions This study provides a proof of concept for detecting liver stage P. vivax and provide the first characterization of P. vivax liver stage forms from an endemic region in India.
Collapse
Affiliation(s)
- Pradeep A Subramani
- National Centre for Biological Sciences (NCBS), Tata Institute of Fundamental Research (TIFR), Bellary Road, Bangalore, 560065, India.,ICMR-National Institute of Malaria Research (NIMR), Indian Council of Medical Research, Bangalore, India
| | - Neha Vartak-Sharma
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), Bangalore, India.,Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advance Studies, Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Seetha Sreekumar
- National Centre for Biological Sciences (NCBS), Tata Institute of Fundamental Research (TIFR), Bellary Road, Bangalore, 560065, India
| | - Pallavi Mathur
- National Centre for Biological Sciences (NCBS), Tata Institute of Fundamental Research (TIFR), Bellary Road, Bangalore, 560065, India
| | - Bhavana Nayer
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), Bangalore, India
| | - Sushrut Dakhore
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), Bangalore, India
| | - Sowmya K Basavanna
- ICMR-National Institute of Malaria Research (NIMR), Indian Council of Medical Research, Bangalore, India
| | - Devaiah M Kalappa
- ICMR-National Institute of Malaria Research (NIMR), Indian Council of Medical Research, Bangalore, India
| | | | - Benudhar Mukhi
- ICMR-National Institute of Malaria Research (NIMR), Indian Council of Medical Research, Bangalore, India
| | - Priyasha Mishra
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), Bangalore, India
| | - Noriko Yoshida
- Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advance Studies, Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Susanta Kumar Ghosh
- ICMR-National Institute of Malaria Research (NIMR), Indian Council of Medical Research, Bangalore, India. .,Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| | | | | | - Brice Campo
- Medicines for Malaria Venture, Geneva, Switzerland
| | - Kouichi Hasegawa
- Institute for Stem Cell Biology and Regenerative Medicine (inStem), Bangalore, India. .,Institute for Integrated Cell-Material Sciences (iCeMS), Institute for Advance Studies, Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| | - Anupkumar R Anvikar
- ICMR-National Institute of Malaria Research (NIMR), Indian Council of Medical Research, New Delhi, India
| | - Neena Valecha
- ICMR-National Institute of Malaria Research (NIMR), Indian Council of Medical Research, New Delhi, India
| | - Varadharajan Sundaramurthy
- National Centre for Biological Sciences (NCBS), Tata Institute of Fundamental Research (TIFR), Bellary Road, Bangalore, 560065, India.
| |
Collapse
|
40
|
Maher SP, Conway AJ, Roth A, Adapa SR, Cualing P, Andolina C, Hsiao J, Turgeon J, Chaumeau V, Johnson M, Palmiotti C, Singh N, Barnes SJ, Patel R, Van Grod V, Carter R, Sun HCS, Sattabongkot J, Campo B, Nosten F, Saadi WM, Adams JH, Jiang RHY, Kyle DE. An adaptable soft-mold embossing process for fabricating optically-accessible, microfeature-based culture systems and application toward liver stage antimalarial compound testing. LAB ON A CHIP 2020; 20:1124-1139. [PMID: 32055808 DOI: 10.1039/c9lc00921c] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Advanced cell culture methods for modeling organ-level structure have been demonstrated to replicate in vivo conditions more accurately than traditional in vitro cell culture. Given that the liver is particularly important to human health, several advanced culture methods have been developed to experiment with liver disease states, including infection with Plasmodium parasites, the causative agent of malaria. These models have demonstrated that intrahepatic parasites require functionally stable hepatocytes to thrive and robust characterization of the parasite populations' response to investigational therapies is dependent on high-content and high-resolution imaging (HC/RI). We previously reported abiotic confinement extends the functional longevity of primary hepatocytes in a microfluidic platform and set out to instill confinement in a microtiter plate platform while maintaining optical accessibility for HC/RI; with an end-goal of producing an improved P. vivax liver stage culture model. We developed a novel fabrication process in which a PDMS soft mold embosses hepatocyte-confining microfeatures into polystyrene, resulting in microfeature-based hepatocyte confinement (μHEP) slides and plates. Our process was optimized to form both microfeatures and culture wells in a single embossing step, resulting in a 100 μm-thick bottom ideal for HC/RI, and was found inexpensively amendable to microfeature design changes. Microfeatures improved intrahepatic parasite infection rates and μHEP systems were used to reconfirm the activity of reference antimalarials in phenotypic dose-response assays. RNAseq of hepatocytes in μHEP systems demonstrated microfeatures sustain hepatic differentiation and function, suggesting broader utility for preclinical hepatic assays; while our tailorable embossing process could be repurposed for developing additional organ models.
Collapse
Affiliation(s)
- Steven P Maher
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA. and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
| | - Amy J Conway
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | - Alison Roth
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | - Swamy R Adapa
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | - Phillip Cualing
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | - Chiara Andolina
- Shoklo Malaria Research Unit, Mahidol Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand & Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - James Hsiao
- Charles Stark Draper Laboratory, Cambridge, MA, USA
| | - Jessica Turgeon
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | - Victor Chaumeau
- Shoklo Malaria Research Unit, Mahidol Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand & Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Myles Johnson
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | | | - Naresh Singh
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | - Samantha J Barnes
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | - Raahil Patel
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | | | - Robert Carter
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | | | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Brice Campo
- Medicines for Malaria Venture, Geneva, Switzerland
| | - François Nosten
- Shoklo Malaria Research Unit, Mahidol Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand & Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - John H Adams
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | - Rays H Y Jiang
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | - Dennis E Kyle
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA. and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
| |
Collapse
|
41
|
Khan HS, Nair VR, Ruhl CR, Alvarez-Arguedas S, Galvan Rendiz JL, Franco LH, Huang L, Shaul PW, Kim J, Xie Y, Mitchell RB, Shiloh MU. Identification of scavenger receptor B1 as the airway microfold cell receptor for Mycobacterium tuberculosis. eLife 2020; 9:52551. [PMID: 32134383 PMCID: PMC7065847 DOI: 10.7554/elife.52551] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 03/04/2020] [Indexed: 12/19/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) can enter the body through multiple routes, including via specialized transcytotic cells called microfold cells (M cell). However, the mechanistic basis for M cell entry remains undefined. Here, we show that M cell transcytosis depends on the Mtb Type VII secretion machine and its major virulence factor EsxA. We identify scavenger receptor B1 (SR-B1) as an EsxA receptor on airway M cells. SR-B1 is required for Mtb binding to and translocation across M cells in mouse and human tissue. Together, our data demonstrate a previously undescribed role for Mtb EsxA in mucosal invasion and identify SR-B1 as the airway M cell receptor for Mtb.
Collapse
Affiliation(s)
- Haaris S Khan
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Vidhya R Nair
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Cody R Ruhl
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Samuel Alvarez-Arguedas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Jorge L Galvan Rendiz
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Luis H Franco
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Linzhang Huang
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Jiwoong Kim
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, United States
| | - Yang Xie
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, United States.,Harold C Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Ron B Mitchell
- Department of Otolaryngology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Michael U Shiloh
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
42
|
Ghartey-Kwansah G, Aboagye B, Adu-Nti F, Opoku YK, Abu EK. Clearing or subverting the enemy: Role of autophagy in protozoan infections. Life Sci 2020; 247:117453. [PMID: 32088215 DOI: 10.1016/j.lfs.2020.117453] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 12/20/2022]
Abstract
The protozoan parasites are evolutionarily divergent, unicellular eukaryotic pathogens representing one of the essential sources of parasitic diseases. These parasites significantly affect the economy and cause public health burdens globally. Protozoan parasites share many cellular features and pathways with their respective host cells. This includes autophagy, a process responsible for self-degradation of the cell's components. There is conservation of the central structural and functional machinery for autophagy in most of the eukaryotic phyla, however, Plasmodium and Toxoplasma possess a decreased number of recognizable autophagy-related proteins (ATG). Plasmodium noticeably lacks clear orthologs of the initiating kinase ATG1/ULK1/2, and both Plasmodium and Toxoplasma lack proteins involved in the nucleation of autophagosomes. These organisms have essential apicoplast, a plastid-like non-photosynthetic organelle, which is an adaptation that is used in penetrating the host cell. Furthermore, available evidence suggests that Leishmania, an intracellular protozoan parasite, induces autophagy in macrophages. The autophagic pathway in Trypanosoma cruzi is activated during metacyclogenesis, a process responsible for the infective forms of parasites. Therefore, numerous pathogens have developed strategies to impair the autophagic mechanism in phagocytes. Regulating autophagy is essential to maintain cellular health as adjustments in the autophagy pathway have been linked to the progression of several physiological and pathological conditions in humans. In this review, we report current advances in autophagy in parasites and their host cells, focusing on the ramifications of these studies in the design of potential anti-protozoan therapeutics.
Collapse
Affiliation(s)
- George Ghartey-Kwansah
- Department of Biomedical Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana; College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Benjamin Aboagye
- Department of Forensic Sciences, College of Agriculture and Natural Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Frank Adu-Nti
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Yeboah Kwaku Opoku
- Department of Biology Education, Faculty of Science, University of Education, Winneba, Ghana
| | - Emmanuel Kwasi Abu
- Department of Optometry and Vision Science, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| |
Collapse
|
43
|
Horta MF, Andrade LO, Martins-Duarte ÉS, Castro-Gomes T. Cell invasion by intracellular parasites - the many roads to infection. J Cell Sci 2020; 133:133/4/jcs232488. [PMID: 32079731 DOI: 10.1242/jcs.232488] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Intracellular parasites from the genera Toxoplasma, Plasmodium, Trypanosoma, Leishmania and from the phylum Microsporidia are, respectively, the causative agents of toxoplasmosis, malaria, Chagas disease, leishmaniasis and microsporidiosis, illnesses that kill millions of people around the globe. Crossing the host cell plasma membrane (PM) is an obstacle these parasites must overcome to establish themselves intracellularly and so cause diseases. The mechanisms of cell invasion are quite diverse and include (1) formation of moving junctions that drive parasites into host cells, as for the protozoans Toxoplasma gondii and Plasmodium spp., (2) subversion of endocytic pathways used by the host cell to repair PM, as for Trypanosoma cruzi and Leishmania, (3) induction of phagocytosis as for Leishmania or (4) endocytosis of parasites induced by specialized structures, such as the polar tubes present in microsporidian species. Understanding the early steps of cell entry is essential for the development of vaccines and drugs for the prevention or treatment of these diseases, and thus enormous research efforts have been made to unveil their underlying biological mechanisms. This Review will focus on these mechanisms and the factors involved, with an emphasis on the recent insights into the cell biology of invasion by these pathogens.
Collapse
Affiliation(s)
- Maria Fátima Horta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Luciana Oliveira Andrade
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Érica Santos Martins-Duarte
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Thiago Castro-Gomes
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| |
Collapse
|
44
|
Vijayan K, Cestari I, Mast FD, Glennon EKK, McDermott SM, Kain HS, Brokaw AM, Aitchison JD, Stuart K, Kaushansky A. Plasmodium Secretion Induces Hepatocyte Lysosome Exocytosis and Promotes Parasite Entry. iScience 2019; 21:603-611. [PMID: 31731198 PMCID: PMC6889558 DOI: 10.1016/j.isci.2019.10.054] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 10/17/2019] [Accepted: 10/24/2019] [Indexed: 12/12/2022] Open
Abstract
The invasion of a suitable host hepatocyte by Plasmodium sporozoites is an essential step in malaria infection. We demonstrate that in infected hepatocytes, lysosomes are redistributed away from the nucleus, and surface exposure of lysosome-associated membrane protein 1 (LAMP1) is increased. Lysosome exocytosis in infected cells occurs independently of sporozoite traversal. Instead, a sporozoite-secreted factor is sufficient for the process. Knockdown of SNARE proteins involved in lysosome-plasma membrane fusion reduces lysosome exocytosis and Plasmodium infection. In contrast, promoting fusion between the lysosome and plasma membrane dramatically increases infection. Our work demonstrates parallels between Plasmodium sporozoite entry of hepatocytes and infection by the excavate pathogen Trypanosoma cruzi and raises the question of whether convergent evolution has shaped host cell invasion by divergent pathogens. Plasmodium sporozoites induce host lysosome exocytosis during invasion Hepatocyte lysosome exocytosis occurs in a SPECT2-independent manner Inhibition of lysosome-plasma membrane fusion inhibits sporozoite invasion Secreted parasite factors are sufficient to induce lysosome exocytosis
Collapse
Affiliation(s)
- Kamalakannan Vijayan
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue, N. Suite 500, Seattle, WA 98109, USA
| | - Igor Cestari
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue, N. Suite 500, Seattle, WA 98109, USA; Institute of Parasitology, McGill University, Ste Anne de Bellevue, QC H9X 3V9, Canada
| | - Fred D Mast
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue, N. Suite 500, Seattle, WA 98109, USA; Institute for Systems Biology, 401 Terry Avenue, N, Seattle, WA 98109, USA
| | - Elizabeth K K Glennon
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue, N. Suite 500, Seattle, WA 98109, USA
| | - Suzanne M McDermott
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue, N. Suite 500, Seattle, WA 98109, USA
| | - Heather S Kain
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue, N. Suite 500, Seattle, WA 98109, USA
| | - Alyssa M Brokaw
- Department of Global Health, University of Washington, Harris Hydraulics Laboratory Box 357965, Seattle, WA 98195-7965, USA
| | - John D Aitchison
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue, N. Suite 500, Seattle, WA 98109, USA; Institute for Systems Biology, 401 Terry Avenue, N, Seattle, WA 98109, USA
| | - Kenneth Stuart
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue, N. Suite 500, Seattle, WA 98109, USA; Department of Global Health, University of Washington, Harris Hydraulics Laboratory Box 357965, Seattle, WA 98195-7965, USA
| | - Alexis Kaushansky
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue, N. Suite 500, Seattle, WA 98109, USA; Department of Global Health, University of Washington, Harris Hydraulics Laboratory Box 357965, Seattle, WA 98195-7965, USA.
| |
Collapse
|
45
|
Affiliation(s)
- Hirdesh Kumar
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Niraj H. Tolia
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
46
|
Mancio-Silva L, Fleming HE, Miller AB, Milstein S, Liebow A, Haslett P, Sepp-Lorenzino L, Bhatia SN. Improving Drug Discovery by Nucleic Acid Delivery in Engineered Human Microlivers. Cell Metab 2019; 29:727-735.e3. [PMID: 30840913 PMCID: PMC6408324 DOI: 10.1016/j.cmet.2019.02.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/21/2018] [Accepted: 02/11/2019] [Indexed: 12/20/2022]
Abstract
The liver plays a central role in metabolism; however, xenobiotic metabolism variations between human hepatocytes and those in model organisms create challenges in establishing functional test beds to detect the potential drug toxicity and efficacy of candidate small molecules. In the emerging areas of RNA interference, viral gene therapy, and genome editing, more robust, long-lasting, and predictive human liver models may accelerate progress. Here, we apply a new modality to a previously established, functionally stable, multi-well bioengineered microliver-fabricated from primary human hepatocytes and supportive stromal cells-in order to advance both small molecule and nucleic acid therapeutic pipelines. Specifically, we achieve robust and durable gene silencing in vitro to tune the human metabolism of small molecules, and demonstrate its capacity to query the potential efficacy and/or toxicity of candidate therapeutics. Additionally, we apply this engineered platform to test siRNAs designed to target hepatocytes and impact human liver genetic and infectious diseases.
Collapse
Affiliation(s)
- Liliana Mancio-Silva
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Heather E Fleming
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Alex B Miller
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Broad Institute, Cambridge, MA 02142, USA
| | - Stuart Milstein
- Alnylam Pharmaceuticals, 300 3rd Street, Cambridge, MA 02142, USA
| | - Abigail Liebow
- Alnylam Pharmaceuticals, 300 3rd Street, Cambridge, MA 02142, USA
| | - Patrick Haslett
- Alnylam Pharmaceuticals, 300 3rd Street, Cambridge, MA 02142, USA
| | | | - Sangeeta N Bhatia
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Broad Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Koch Institute for Integrative Cancer Research, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
47
|
Tweedell RE, Tao D, Hamerly T, Robinson TM, Larsen S, Grønning AGB, Norris AM, King JG, Law HCH, Baumbach J, Bergmann-Leitner ES, Dinglasan RR. The Selection of a Hepatocyte Cell Line Susceptible to Plasmodium falciparum Sporozoite Invasion That Is Associated With Expression of Glypican-3. Front Microbiol 2019; 10:127. [PMID: 30891005 PMCID: PMC6413710 DOI: 10.3389/fmicb.2019.00127] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 01/21/2019] [Indexed: 01/09/2023] Open
Abstract
In vitro studies of liver stage (LS) development of the human malaria parasite Plasmodium falciparum are technically challenging; therefore, fundamental questions about hepatocyte receptors for invasion that can be targeted to prevent infection remain unanswered. To identify novel receptors and to further understand human hepatocyte susceptibility to P. falciparum sporozoite invasion, we created an optimized in vitro system by mimicking in vivo liver conditions and using the subcloned HC-04.J7 cell line that supports mean infection rates of 3-5% and early development of P. falciparum exoerythrocytic forms-a 3- to 5-fold improvement on current in vitro hepatocarcinoma models for P. falciparum invasion. We juxtaposed this invasion-susceptible cell line with an invasion-resistant cell line (HepG2) and performed comparative proteomics and RNA-seq analyses to identify host cell surface molecules and pathways important for sporozoite invasion of host cells. We identified and investigated a hepatocyte cell surface heparan sulfate proteoglycan, glypican-3, as a putative mediator of sporozoite invasion. We also noted the involvement of pathways that implicate the importance of the metabolic state of the hepatocyte in supporting LS development. Our study highlights important features of hepatocyte biology, and specifically the potential role of glypican-3, in mediating P. falciparum sporozoite invasion. Additionally, it establishes a simple in vitro system to study the LS with improved invasion efficiency. This work paves the way for the greater malaria and liver biology communities to explore fundamental questions of hepatocyte-pathogen interactions and extend the system to other human malaria parasite species, like P. vivax.
Collapse
Affiliation(s)
- Rebecca E Tweedell
- Department of Infectious Diseases and Immunology, Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States.,W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Dingyin Tao
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Timothy Hamerly
- Department of Infectious Diseases and Immunology, Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States.,W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Tanisha M Robinson
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Simon Larsen
- Computational BioMedicine Lab, Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
| | - Alexander G B Grønning
- Computational BioMedicine Lab, Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
| | - Alessandra M Norris
- Department of Infectious Diseases and Immunology, Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
| | - Jonas G King
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States.,Department of Biochemistry, Molecular Biology, Entomology & Plant Pathology, Mississippi State University, Starkville, MS, United States
| | - Henry Chun Hin Law
- Department of Infectious Diseases and Immunology, Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
| | - Jan Baumbach
- Computational BioMedicine Lab, Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark.,Chair of Experimental Bioinformatics, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | - Elke S Bergmann-Leitner
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Rhoel R Dinglasan
- Department of Infectious Diseases and Immunology, Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States.,W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
48
|
Dundas K, Shears MJ, Sinnis P, Wright GJ. Important Extracellular Interactions between Plasmodium Sporozoites and Host Cells Required for Infection. Trends Parasitol 2018; 35:129-139. [PMID: 30583849 DOI: 10.1016/j.pt.2018.11.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 11/22/2018] [Accepted: 11/26/2018] [Indexed: 01/08/2023]
Abstract
Malaria is an infectious disease, caused by Plasmodium parasites, that remains a major global health problem. Infection begins when salivary gland sporozoites are transmitted through the bite of an infected mosquito. Once within the host, sporozoites navigate through the dermis, into the bloodstream, and eventually invade hepatocytes. While we have an increasingly sophisticated cellular description of this journey, our molecular understanding of the extracellular interactions between the sporozoite and mammalian host that regulate migration and invasion remain comparatively poor. Here, we review the current state of our understanding, highlight the technical limitations that have frustrated progress, and outline how new approaches will help to address this knowledge gap with the ultimate aim of improving malaria treatments.
Collapse
Affiliation(s)
- Kirsten Dundas
- Cell Surface Signalling Laboratory and Parasites and Microbes Programme, Wellcome Trust Sanger Institute, Cambridge, CB10 1SA, UK
| | - Melanie J Shears
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Photini Sinnis
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Gavin J Wright
- Cell Surface Signalling Laboratory and Parasites and Microbes Programme, Wellcome Trust Sanger Institute, Cambridge, CB10 1SA, UK.
| |
Collapse
|
49
|
Arredondo SA, Swearingen KE, Martinson T, Steel R, Dankwa DA, Harupa A, Camargo N, Betz W, Vigdorovich V, Oliver BG, Kangwanrangsan N, Ishino T, Sather N, Mikolajczak S, Vaughan AM, Torii M, Moritz RL, Kappe SHI. The Micronemal Plasmodium Proteins P36 and P52 Act in Concert to Establish the Replication-Permissive Compartment Within Infected Hepatocytes. Front Cell Infect Microbiol 2018; 8:413. [PMID: 30547015 PMCID: PMC6280682 DOI: 10.3389/fcimb.2018.00413] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 11/08/2018] [Indexed: 12/15/2022] Open
Abstract
Within the liver, Plasmodium sporozoites traverse cells searching for a "suitable" hepatocyte, invading these cells through a process that results in the formation of a parasitophorous vacuole (PV), within which the parasite undergoes intracellular replication as a liver stage. It was previously established that two members of the Plasmodium s48/45 protein family, P36 and P52, are essential for productive invasion of host hepatocytes by sporozoites as their simultaneous deletion results in growth-arrested parasites that lack a PV. Recent studies point toward a pathway of entry possibly involving the interaction of P36 with hepatocyte receptors EphA2, CD81, and SR-B1. However, the relationship between P36 and P52 during sporozoite invasion remains unknown. Here we show that parasites with a single P52 or P36 gene deletion each lack a PV after hepatocyte invasion, thereby pheno-copying the lack of a PV observed for the P52/P36 dual gene deletion parasite line. This indicates that both proteins are equally important in the establishment of a PV and act in the same pathway. We created a Plasmodium yoelii P36mCherry tagged parasite line that allowed us to visualize the subcellular localization of P36 and found that it partially co-localizes with P52 in the sporozoite secretory microneme organelles. Furthermore, through co-immunoprecipitation studies in vivo, we determined that P36 and P52 form a protein complex in sporozoites, indicating a concerted function for both proteins within the PV formation pathway. However, upon sporozoite stimulation, only P36 was released as a secreted protein while P52 was not. Our results support a model in which the putatively glycosylphosphatidylinositol (GPI)-anchored P52 may serve as a scaffold to facilitate the interaction of secreted P36 with the host cell during sporozoite invasion of hepatocytes.
Collapse
Affiliation(s)
- Silvia A. Arredondo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | | | - Thomas Martinson
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Ryan Steel
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Dorender A. Dankwa
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Anke Harupa
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Nelly Camargo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - William Betz
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Vladimir Vigdorovich
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Brian G. Oliver
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Niwat Kangwanrangsan
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Tomoko Ishino
- Department of Molecular Parasitology, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Japan
| | - Noah Sather
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Sebastian Mikolajczak
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Ashley M. Vaughan
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Motomi Torii
- Department of Molecular Parasitology, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Japan
| | | | - Stefan H. I. Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| |
Collapse
|
50
|
Hepatitis C virus enters liver cells using the CD81 receptor complex proteins calpain-5 and CBLB. PLoS Pathog 2018; 14:e1007111. [PMID: 30024968 PMCID: PMC6053247 DOI: 10.1371/journal.ppat.1007111] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 05/18/2018] [Indexed: 12/24/2022] Open
Abstract
Hepatitis C virus (HCV) and the malaria parasite Plasmodium use the membrane protein CD81 to invade human liver cells. Here we mapped 33 host protein interactions of CD81 in primary human liver and hepatoma cells using high-resolution quantitative proteomics. In the CD81 protein network, we identified five proteins which are HCV entry factors or facilitators including epidermal growth factor receptor (EGFR). Notably, we discovered calpain-5 (CAPN5) and the ubiquitin ligase Casitas B-lineage lymphoma proto-oncogene B (CBLB) to form a complex with CD81 and support HCV entry. CAPN5 and CBLB were required for a post-binding and pre-replication step in the HCV life cycle. Knockout of CAPN5 and CBLB reduced susceptibility to all tested HCV genotypes, but not to other enveloped viruses such as vesicular stomatitis virus and human coronavirus. Furthermore, Plasmodium sporozoites relied on a distinct set of CD81 interaction partners for liver cell entry. Our findings reveal a comprehensive CD81 network in human liver cells and show that HCV and Plasmodium highjack selective CD81 interactions, including CAPN5 and CBLB for HCV, to invade cells. CD81 is a cell membrane protein, which functions as entry factor for hepatitis C virus (HCV) and malaria sporozoites in the human liver. Currently, it remains enigmatic how CD81 guides the entry process of both pathogens and whether it functions in a similar way during liver cell invasion of HCV and malaria parasites. Here, we use high resolution quantitative proteomics to identify CD81 associated host proteins in liver cells. We found that at least 33 proteins form a complex with CD81, 23 of which were not reported as interaction partners before. We further determined that at least five CD81 interactors are HCV host factors, among them calpain-5 (CAPN5) and the ubiquitin ligase Casitas B-lineage lymphoma proto-oncogene B (CBLB). All tested HCV genotypes require CAPN5 and CBLB for full infection, but neither malaria parasites nor other tested enveloped virus rely on CAPN5 or CBLB. Our study maps the liver cell interactome of CD81 and provides new insight into the distinct cell invasion mechanisms of HCV and malaria parasites.
Collapse
|