1
|
Goswami D, Arredondo SA, Betz W, Armstrong J, Kumar S, Zanghi G, Patel H, Camargo N, Oualim KMZ, Seilie AM, Schneider S, Murphy SC, Kappe SHI, Vaughan AM. A conserved Plasmodium nuclear protein is critical for late liver stage development. Commun Biol 2024; 7:1387. [PMID: 39455824 PMCID: PMC11511937 DOI: 10.1038/s42003-024-07063-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Malaria, caused by Plasmodium parasites, imposes a significant health burden and live-attenuated parasites are being pursued as vaccines. Here, we report on the creation of a genetically attenuated parasite by the deletion of Plasmodium LINUP, encoding a liver stage nuclear protein. In the rodent parasite Plasmodium yoelii, LINUP expression was restricted to liver stage nuclei after the onset of liver stage schizogony. Compared to wildtype P. yoelii, P. yoelii LINUP gene deletion parasites (linup-) exhibited no phenotype in blood stages and mosquito stages but suffered developmental arrest late in liver stage schizogony with a pronounced defect in exo-erythrocytic merozoite formation. This defect caused severe attenuation of the liver stage-to-blood stage transition and immunization of mice with linup - parasites conferred robust protection against infectious sporozoite challenge. LINUP gene deletion in the human parasite Plasmodium falciparum also caused a severe defect in late liver stage differentiation. Importantly, P. falciparum linup - liver stages completely failed to transition from the liver stage to a viable blood stage infection in a humanized mouse model. These results suggest that P. falciparum LINUP is an ideal target for late liver stage attenuation that can be incorporated into a late liver stage-arresting replication competent whole parasite vaccine.
Collapse
Affiliation(s)
- Debashree Goswami
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA.
| | - Silvia A Arredondo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - William Betz
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Janna Armstrong
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Sudhir Kumar
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Gigliola Zanghi
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Hardik Patel
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nelly Camargo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Kenza M Z Oualim
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Annette M Seilie
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Sophia Schneider
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Sean C Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, 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.
| | - Ashley M Vaughan
- Center for Global Infectious Disease Research, 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
|
2
|
Targetome Analysis of Malaria Sporozoite Transcription Factor AP2-Sp Reveals Its Role as a Master Regulator. mBio 2023; 14:e0251622. [PMID: 36622145 PMCID: PMC9973277 DOI: 10.1128/mbio.02516-22] [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] [Indexed: 01/10/2023] Open
Abstract
Malaria transmission to humans begins with sporozoite infection of the liver. The elucidation of gene regulation during the sporozoite stage will promote the investigation of mechanisms of liver infection by this parasite and contribute to the development of strategies for preventing malaria transmission. AP2-Sp is a transcription factor (TF) essential for the formation of sporozoites or sporogony, which takes place in oocysts in the midguts of infected mosquitoes. To understand the role of this TF in the transcriptional regulatory system of this stage, we performed chromatin immunoprecipitation sequencing (ChIP-seq) analyses using whole mosquito midguts containing late oocysts as starting material and explored its genome-wide target genes. We identified 697 target genes, comprising those involved in distinct processes parasites experience during this stage, from sporogony to development into the liver stage and representing the majority of genes highly expressed in the sporozoite stage. These results suggest that AP2-Sp determines basal patterns of gene expression by targeting a broad range of genes directly. The ChIP-seq analyses also showed that AP2-Sp maintains its own expression by a transcriptional autoactivation mechanism (positive-feedback loop) and induces all TFs reported to be transcribed at this stage, including AP2-Sp2, AP2-Sp3, and SLARP. The results showed that AP2-Sp exists at the top of the transcriptional cascade of this stage and triggers the formation of this stage as a master regulator. IMPORTANCE The sporozoite stage plays a central role in malaria transmission from a mosquito to vertebrate host and is an important target for antimalarial strategies. AP2-Sp is a candidate master transcription factor for the sporozoite stage. However, study of its role in gene regulation has been hampered because of difficulties in performing genome-wide studies of gene regulation in this stage. Here, we conquered this problem and revealed that AP2-Sp has the following prominent features as a master transcription factor. First, it determines the repertory of gene expression during this stage. Second, it maintains its own expression through a transcriptional positive-feedback loop and induces all other transcription factors specifically expressed in this stage. This study represents a major breakthrough in fully understanding gene regulation in this important malarial stage.
Collapse
|
3
|
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
|
4
|
Richie TL, Church LWP, Murshedkar T, Billingsley PF, James ER, Chen MC, Abebe Y, KC N, Chakravarty S, Dolberg D, Healy SA, Diawara H, Sissoko MS, Sagara I, Cook DM, Epstein JE, Mordmüller B, Kapulu M, Kreidenweiss A, Franke-Fayard B, Agnandji ST, López Mikue MSA, McCall MBB, Steinhardt L, Oneko M, Olotu A, Vaughan AM, Kublin JG, Murphy SC, Jongo S, Tanner M, Sirima SB, Laurens MB, Daubenberger C, Silva JC, Lyke KE, Janse CJ, Roestenberg M, Sauerwein RW, Abdulla S, Dicko A, Kappe SHI, Lee Sim BK, Duffy PE, Kremsner PG, Hoffman SL. Sporozoite immunization: innovative translational science to support the fight against malaria. Expert Rev Vaccines 2023; 22:964-1007. [PMID: 37571809 PMCID: PMC10949369 DOI: 10.1080/14760584.2023.2245890] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 08/04/2023] [Indexed: 08/13/2023]
Abstract
INTRODUCTION Malaria, a devastating febrile illness caused by protozoan parasites, sickened 247,000,000 people in 2021 and killed 619,000, mostly children and pregnant women in sub-Saharan Africa. A highly effective vaccine is urgently needed, especially for Plasmodium falciparum (Pf), the deadliest human malaria parasite. AREAS COVERED Sporozoites (SPZ), the parasite stage transmitted by Anopheles mosquitoes to humans, are the only vaccine immunogen achieving >90% efficacy against Pf infection. This review describes >30 clinical trials of PfSPZ vaccines in the U.S.A., Europe, Africa, and Asia, based on first-hand knowledge of the trials and PubMed searches of 'sporozoites,' 'malaria,' and 'vaccines.' EXPERT OPINION First generation (radiation-attenuated) PfSPZ vaccines are safe, well tolerated, 80-100% efficacious against homologous controlled human malaria infection (CHMI) and provide 18-19 months protection without boosting in Africa. Second generation chemo-attenuated PfSPZ are more potent, 100% efficacious against stringent heterologous (variant strain) CHMI, but require a co-administered drug, raising safety concerns. Third generation, late liver stage-arresting, replication competent (LARC), genetically-attenuated PfSPZ are expected to be both safe and highly efficacious. Overall, PfSPZ vaccines meet safety, tolerability, and efficacy requirements for protecting pregnant women and travelers exposed to Pf in Africa, with licensure for these populations possible within 5 years. Protecting children and mass vaccination programs to block transmission and eliminate malaria are long-term objectives.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Sara A. Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Halimatou Diawara
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Mahamadou S. Sissoko
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Issaka Sagara
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - David M. Cook
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Judith E. Epstein
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin Mordmüller
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Melissa Kapulu
- Biosciences Department, Kenya Medical Research Institute KEMRI-Wellcome Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Andrea Kreidenweiss
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), partner site Tübingen, Tübingen, Germany
| | | | - Selidji T. Agnandji
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | | | - Matthew B. B. McCall
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Laura Steinhardt
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Martina Oneko
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Ally Olotu
- Bagamoyo Research and Training Center, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Ashley M. Vaughan
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - James G. Kublin
- Department of Global Health, University of Washington, Seattle, WA, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sean C. Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases and Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Said Jongo
- Bagamoyo Research and Training Center, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Marcel Tanner
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | | | - Matthew B. Laurens
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Claudia Daubenberger
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Joana C. Silva
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kirsten E. Lyke
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Chris J. Janse
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Meta Roestenberg
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert W. Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Salim Abdulla
- Bagamoyo Research and Training Center, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Alassane Dicko
- Malaria Research and Training Center, Mali-NIAID ICER, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Stefan H. I. Kappe
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Patrick E. Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter G. Kremsner
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), partner site Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | | |
Collapse
|
5
|
A member of the tryptophan-rich protein family is required for efficient sequestration of Plasmodium berghei schizonts. PLoS Pathog 2022; 18:e1010846. [PMID: 36126089 PMCID: PMC9524624 DOI: 10.1371/journal.ppat.1010846] [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: 07/15/2022] [Revised: 09/30/2022] [Accepted: 08/31/2022] [Indexed: 11/20/2022] Open
Abstract
Protein export and host membrane remodeling are crucial for multiple Plasmodium species to establish a niche in infected hosts. To better understand the contribution of these processes to successful parasite infection in vivo, we sought to find and characterize protein components of the intraerythrocytic Plasmodium berghei-induced membrane structures (IBIS) that form in the cytoplasm of infected erythrocytes. We identified proteins that immunoprecipitate with IBIS1, a signature member of the IBIS in P. berghei-infected erythrocytes. In parallel, we also report our data describing proteins that co-precipitate with the PTEX (Plasmodium translocon of exported proteins) component EXP2. To validate our findings, we examined the location of three candidate IBIS1-interactors that are conserved across multiple Plasmodium species, and we found they localized to IBIS in infected red blood cells and two further colocalized with IBIS1 in the liver-stage parasitophorous vacuole membrane. Successful gene deletion revealed that these two tryptophan-rich domain-containing proteins, termed here IPIS2 and IPIS3 (for intraerythrocytic Plasmodium-induced membrane structures), are required for efficient blood-stage growth. Erythrocytes infected with IPIS2-deficient schizonts in particular fail to bind CD36 as efficiently as wild-type P. berghei-infected cells and therefore fail to effectively sequester out of the circulating blood. Our findings support the idea that intra-erythrocytic membrane compartments are required across species for alterations of the host erythrocyte that facilitate interactions of infected cells with host tissues.
Collapse
|
6
|
Kreutzfeld O, Grützke J, Ingmundson A, Müller K, Matuschewski K. Absence of PEXEL-Dependent Protein Export in Plasmodium Liver Stages Cannot Be Restored by Gain of the HSP101 Protein Translocon ATPase. Front Genet 2021; 12:742153. [PMID: 34956312 PMCID: PMC8693896 DOI: 10.3389/fgene.2021.742153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
Host cell remodeling is critical for successful Plasmodium replication inside erythrocytes and achieved by targeted export of parasite-encoded proteins. In contrast, during liver infection the malarial parasite appears to avoid protein export, perhaps to limit exposure of parasite antigens by infected liver cells. HSP101, the force-generating ATPase of the protein translocon of exported proteins (PTEX) is the only component that is switched off during early liver infection. Here, we generated transgenic Plasmodium berghei parasite lines that restore liver stage expression of HSP101. HSP101 expression in infected hepatocytes was achieved by swapping the endogenous promoter with the ptex150 promoter and by inserting an additional copy under the control of the elongation one alpha (ef1α) promoter. Both promoters drive constitutive and, hence, also pre-erythrocytic expression. Transgenic parasites were able to complete the life cycle, but failed to export PEXEL-proteins in early liver stages. Our results suggest that PTEX-dependent early liver stage export cannot be restored by addition of HSP101, indicative of alternative export complexes or other functions of the PTEX core complex during liver infection.
Collapse
Affiliation(s)
- Oriana Kreutzfeld
- Molecular Parasitology, Institute of Biology/Faculty for Life Sciences, Humboldt Universität zu Berlin, Berlin, Germany.,Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany.,Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Josephine Grützke
- Molecular Parasitology, Institute of Biology/Faculty for Life Sciences, Humboldt Universität zu Berlin, Berlin, Germany.,Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany.,Department of Biological Safety, Federal Institute for Risk Assessment, Berlin, Germany
| | - Alyssa Ingmundson
- Molecular Parasitology, Institute of Biology/Faculty for Life Sciences, Humboldt Universität zu Berlin, Berlin, Germany.,Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Katja Müller
- Molecular Parasitology, Institute of Biology/Faculty for Life Sciences, Humboldt Universität zu Berlin, Berlin, Germany.,Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Kai Matuschewski
- Molecular Parasitology, Institute of Biology/Faculty for Life Sciences, Humboldt Universität zu Berlin, Berlin, Germany.,Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
7
|
Brandsma AM, Hilmer C, Rauch M, Matuschewski K, Montagna GN. Plasmodium early transcribed membrane proteins appear tailored to the host range of malaria parasites. Int J Parasitol 2021; 52:135-143. [PMID: 34715088 DOI: 10.1016/j.ijpara.2021.08.005] [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: 04/15/2021] [Revised: 08/11/2021] [Accepted: 08/18/2021] [Indexed: 11/05/2022]
Abstract
Early transcribed membrane proteins form a unique protein family in malaria parasites. These molecules are expressed during Plasmodium intracellular phases and inserted at the parasite parasitophorus vacuole membrane, which constitutes the host-parasite interface. Upregulated in infectious sporozoites 4 (UIS4) is an essential early transcribed membrane protein of liver stages of the murine malaria model parasite Plasmodium berghei. Despite its relevance for liver stage maturation, the molecular functions of UIS4 remain elusive, and UIS4 orthologs in human malaria parasites have not yet been identified. In order to characterise functional domains of UIS4, we generated P. berghei parasites carrying a carboxy-terminally truncated version of UIS4. We observed that uis4Δc parasites are severely impaired in liver stage development, similar to uis4(-) parasites, indicating an important role of the C-terminal domain for UIS4 function. To test whether members of the P. falciparum early transcribed membrane protein family are potential UIS4 orthologs, we selected candidates based on structural homology and parasitophorous vacuole membrane localization. We generated transgenic P. berghei parasites where UIS4 was replaced by Plasmodium falciparum ETRAMP8 or ETRAMP10.3. Both early transcribed membrane proteins were expressed in transgenic parasite lines, but liver stage maturation was impaired, indicating that the selected early transcribed membrane proteins failed to substitute the function of UIS4. As a control, we included the UIS4 ortholog from the murine parasite Plasmodium chaubaudi. We observed that PcUIS4 successfully restores UIS4 function in P. berghei. Together, these results suggest that Plasmodium parasites express tailor-made parasitophorous vacuole membrane proteins that might at least partially explain the narrow host range of malaria parasites.
Collapse
Affiliation(s)
- Arianne M Brandsma
- Parasitology Unit, Max Planck Institute for Infection Biology, 10117 Berlin, Germany; Princess Máxima Center for Pediatric Oncology, Heidelberg 25, 3584 CS Utrecht, The Netherlands
| | - Cecilie Hilmer
- Parasitology Unit, Max Planck Institute for Infection Biology, 10117 Berlin, Germany
| | - Manuel Rauch
- Parasitology Unit, Max Planck Institute for Infection Biology, 10117 Berlin, Germany; Dept. of Molecular Parasitology, Institute of Biology, Humboldt University, 10115 Berlin, Germany
| | - Kai Matuschewski
- Parasitology Unit, Max Planck Institute for Infection Biology, 10117 Berlin, Germany; Dept. of Molecular Parasitology, Institute of Biology, Humboldt University, 10115 Berlin, Germany
| | - Georgina N Montagna
- Parasitology Unit, Max Planck Institute for Infection Biology, 10117 Berlin, Germany; Instituto de Investigaciones Biotecnológicas Dr Rodolfo Ugalde' (IIBio), UNSAM-CONICET 1650 San Martín, Buenos Aires, Argentina.
| |
Collapse
|
8
|
Scheunemann JF, Reichwald JJ, Korir PJ, Kuehlwein JM, Jenster LM, Hammerschmidt-Kamper C, Lewis MD, Klocke K, Borsche M, Schwendt KE, Soun C, Thiebes S, Limmer A, Engel DR, Mueller AK, Hoerauf A, Hübner MP, Schumak B. Eosinophils Suppress the Migration of T Cells Into the Brain of Plasmodium berghei-Infected Ifnar1-/- Mice and Protect Them From Experimental Cerebral Malaria. Front Immunol 2021; 12:711876. [PMID: 34659202 PMCID: PMC8514736 DOI: 10.3389/fimmu.2021.711876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/13/2021] [Indexed: 11/29/2022] Open
Abstract
Cerebral malaria is a potentially lethal disease, which is caused by excessive inflammatory responses to Plasmodium parasites. Here we use a newly developed transgenic Plasmodium berghei ANKA (PbAAma1OVA) parasite that can be used to study parasite-specific T cell responses. Our present study demonstrates that Ifnar1-/- mice, which lack type I interferon receptor-dependent signaling, are protected from experimental cerebral malaria (ECM) when infected with this novel parasite. Although CD8+ T cell responses generated in the spleen are essential for the development of ECM, we measured comparable parasite-specific cytotoxic T cell responses in ECM-protected Ifnar1-/- mice and wild type mice suffering from ECM. Importantly, CD8+ T cells were increased in the spleens of ECM-protected Ifnar1-/- mice and the blood-brain-barrier remained intact. This was associated with elevated splenic levels of CCL5, a T cell and eosinophil chemotactic chemokine, which was mainly produced by eosinophils, and an increase in eosinophil numbers. Depletion of eosinophils enhanced CD8+ T cell infiltration into the brain and increased ECM induction in PbAAma1OVA-infected Ifnar1-/- mice. However, eosinophil-depletion did not reduce the CD8+ T cell population in the spleen or reduce splenic CCL5 concentrations. Our study demonstrates that eosinophils impact CD8+ T cell migration and proliferation during PbAAma1OVA-infection in Ifnar1-/- mice and thereby are contributing to the protection from ECM.
Collapse
Affiliation(s)
- Johanna F Scheunemann
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Julia J Reichwald
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Patricia Jebett Korir
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Janina M Kuehlwein
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Lea-Marie Jenster
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | | | - Matthew D Lewis
- Parasitology Unit, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Katrin Klocke
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Max Borsche
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Kim E Schwendt
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Camille Soun
- Institute for Experimental Immunology and Imaging, University Hospital Essen, Essen, Germany
| | - Stephanie Thiebes
- Institute for Experimental Immunology and Imaging, University Hospital Essen, Essen, Germany
| | - Andreas Limmer
- Clinic for Anesthesiology and Intensive Care, University Hospital Essen, Essen, Germany
| | - Daniel R Engel
- Institute for Experimental Immunology and Imaging, University Hospital Essen, Essen, Germany
| | - Ann-Kristin Mueller
- Parasitology Unit, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Infection Research (DZIF), Heidelberg, Germany
| | - Achim Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany.,German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Marc P Hübner
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany.,German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Beatrix Schumak
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
9
|
Sekar V, Rivero A, Pigeault R, Gandon S, Drews A, Ahren D, Hellgren O. Gene regulation of the avian malaria parasite Plasmodium relictum, during the different stages within the mosquito vector. Genomics 2021; 113:2327-2337. [PMID: 34023365 DOI: 10.1016/j.ygeno.2021.05.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/26/2021] [Accepted: 05/18/2021] [Indexed: 10/21/2022]
Abstract
The malaria parasite Plasmodium relictum is one of the most widespread species of avian malaria. As in the case of its human counterparts, bird Plasmodium undergoes a complex life cycle infecting two hosts: the arthropod vector and the vertebrate host. In this study, we examined transcriptomes of P. relictum (SGS1) during crucial timepoints within its vector, Culex pipiens quinquefasciatus. Differential gene-expression analyses identified genes linked to the parasites life-stages at: i) a few minutes after the blood meal is ingested, ii) during peak oocyst production phase, iii) during peak sporozoite phase and iv) during the late-stages of the infection. A large amount of genes coding for functions linked to host-immune invasion and multifunctional genes was active throughout the infection cycle. One gene associated with a conserved Plasmodium membrane protein with unknown function was upregulated throughout the parasite development in the vector, suggesting an important role in the successful completion of the sporogonic cycle. Gene expression analysis further identified genes, with unknown functions to be significantly differentially expressed during the infection in the vector as well as upregulation of reticulocyte-binding proteins, which raises the possibility of the multifunctionality of these RBPs. We establish the existence of highly stage-specific pathways being overexpressed during the infection. This first study of gene-expression of a non-human Plasmodium species in its vector provides a comprehensive insight into the molecular mechanisms of the common avian malaria parasite P. relictum and provides essential information on the evolutionary diversity in gene regulation of the Plasmodium's vector stages.
Collapse
Affiliation(s)
| | - Ana Rivero
- MIVEGEC (CNRS - Université de Montpellier - IRD), 34394 Montpellier, France; CREES (Centre de Recherche en Ecologie et Evolution de la Santé), 34394 Montpellier, France
| | - Romain Pigeault
- Department of Biology, Lund University, Sweden; Department of Ecology and Evolution, CH-1015 Lausanne, Switzerland
| | - Sylvain Gandon
- CEFE (CNRS - Université de Montpellier - Université Paul-Valéry - EPHE - IRD), Montpellier, France
| | - Anna Drews
- MEMEG, Department of Biology, Lund University, Sweden
| | - Dag Ahren
- National Bioinformatics Infrastructure Sweden (NBIS), SciLifeLab, Department of Biology, Lund, Sweden
| | - Olof Hellgren
- MEMEG, Department of Biology, Lund University, Sweden.
| |
Collapse
|
10
|
Novel insights from the Plasmodium falciparum sporozoite-specific proteome by probabilistic integration of 26 studies. PLoS Comput Biol 2021; 17:e1008067. [PMID: 33930021 PMCID: PMC8115857 DOI: 10.1371/journal.pcbi.1008067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 05/12/2021] [Accepted: 04/06/2021] [Indexed: 11/19/2022] Open
Abstract
Plasmodium species, the causative agent of malaria, have a complex life cycle involving two hosts. The sporozoite life stage is characterized by an extended phase in the mosquito salivary glands followed by free movement and rapid invasion of hepatocytes in the human host. This transmission stage has been the subject of many transcriptomics and proteomics studies and is also targeted by the most advanced malaria vaccine. We applied Bayesian data integration to determine which proteins are not only present in sporozoites but are also specific to that stage. Transcriptomic and proteomic Plasmodium data sets from 26 studies were weighted for how representative they are for sporozoites, based on a carefully assembled gold standard for Plasmodium falciparum (Pf) proteins known to be present or absent during the sporozoite life stage. Of 5418 Pf genes for which expression data were available at the RNA level or at the protein level, 975 were identified as enriched in sporozoites and 90 specific to them. We show that Pf sporozoites are enriched for proteins involved in type II fatty acid synthesis in the apicoplast and GPI anchor synthesis, but otherwise appear metabolically relatively inactive in the salivary glands of mosquitos. Newly annotated hypothetical sporozoite-specific and sporozoite-enriched proteins highlight sporozoite-specific functions. They include PF3D7_0104100 that we identified to be homologous to the prominin family, which in human has been related to a quiescent state of cancer cells. We document high levels of genetic variability for sporozoite proteins, specifically for sporozoite-specific proteins that elicit antibodies in the human host. Nevertheless, we can identify nine relatively well-conserved sporozoite proteins that elicit antibodies and that together can serve as markers for previous exposure. Our understanding of sporozoite biology benefits from identifying key pathways that are enriched during this life stage. This work can guide studies of molecular mechanisms underlying sporozoite biology and potential well-conserved targets for marker and drug development. When a person is bitten by an infectious malaria mosquito, sporozoites are injected into the skin with mosquito saliva. These sporozoites then travel to the liver, invade hepatocytes and multiply before the onset of the symptom-causing blood stage of malaria. By integrating published data, we contrast sporozoite protein expression with other life stages to filter out the unique features of sporozoites that help us understand this stage. We used a “guideline” that we derived from the literature on individual proteins so that we knew which proteins should be present or absent at the sporozoite stage, allowing us to weigh 26 data sets for their relevance to sporozoites. Among the newly discovered sporozoite-specific genes are candidates for fatty acid synthesis while others might play a role keeping the sporozoites in an inactive state in the mosquito salivary glands. Furthermore, we show that most sporozoite-specific proteins are genetically more variable than non-sporozoite proteins. We identify a set of conserved sporozoite proteins against which antibodies can serve as markers of recent exposure to sporozoites or that can serve as vaccine candidates. Our predictions of sporozoite-specific proteins and the assignment of previously unknown functions give new insights into the biology of this life stage.
Collapse
|
11
|
Müller K, Silvie O, Mollenkopf HJ, Matuschewski K. Pleiotropic Roles for the Plasmodium berghei RNA Binding Protein UIS12 in Transmission and Oocyst Maturation. Front Cell Infect Microbiol 2021; 11:624945. [PMID: 33747980 PMCID: PMC7973279 DOI: 10.3389/fcimb.2021.624945] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 01/15/2021] [Indexed: 11/21/2022] Open
Abstract
Colonization of the mosquito host by Plasmodium parasites is achieved by sexually differentiated gametocytes. Gametocytogenesis, gamete formation and fertilization are tightly regulated processes, and translational repression is a major regulatory mechanism for stage conversion. Here, we present a characterization of a Plasmodium berghei RNA binding protein, UIS12, that contains two conserved eukaryotic RNA recognition motifs (RRM). Targeted gene deletion resulted in viable parasites that replicate normally during blood infection, but form fewer gametocytes. Upon transmission to Anopheles stephensi mosquitoes, both numbers and size of midgut-associated oocysts were reduced and their development stopped at an early time point. As a consequence, no salivary gland sporozoites were formed indicative of a complete life cycle arrest in the mosquito vector. Comparative transcript profiling in mutant and wild-type infected red blood cells revealed a decrease in transcript abundance of mRNAs coding for signature gamete-, ookinete-, and oocyst-specific proteins in uis12(-) parasites. Together, our findings indicate multiple roles for UIS12 in regulation of gene expression after blood infection in good agreement with the pleiotropic defects that terminate successful sporogony and onward transmission to a new vertebrate host.
Collapse
Affiliation(s)
- Katja Müller
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany.,Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Olivier Silvie
- Centre d'Immunologie et des Maladies Infectieuses, INSERM, CNRS, Sorbonne Université, Paris, France
| | - Hans-Joachim Mollenkopf
- Core Facility Microarray/Genomics, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Kai Matuschewski
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany.,Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
12
|
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: 4.3] [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
|
13
|
Briquet S, Marinach C, Silvie O, Vaquero C. Preparing for Transmission: Gene Regulation in Plasmodium Sporozoites. Front Cell Infect Microbiol 2021; 10:618430. [PMID: 33585284 PMCID: PMC7878544 DOI: 10.3389/fcimb.2020.618430] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/16/2020] [Indexed: 11/13/2022] Open
Abstract
Plasmodium sporozoites are transmitted to mammals by anopheline mosquitoes and first infect the liver, where they transform into replicative exoerythrocytic forms, which subsequently release thousands of merozoites that invade erythrocytes and initiate the malaria disease. In some species, sporozoites can transform into dormant hypnozoites in the liver, which cause malaria relapses upon reactivation. Transmission from the insect vector to a mammalian host is a critical step of the parasite life cycle, and requires tightly regulated gene expression. Sporozoites are formed inside oocysts in the mosquito midgut and become fully infectious after colonization of the insect salivary glands, where they remain quiescent until transmission. Parasite maturation into infectious sporozoites is associated with reprogramming of the sporozoite transcriptome and proteome, which depends on multiple layers of transcriptional and post-transcriptional regulatory mechanisms. An emerging scheme is that gene expression in Plasmodium sporozoites is controlled by alternating waves of transcription activity and translational repression, which shape the parasite RNA and protein repertoires for successful transition from the mosquito vector to the mammalian host.
Collapse
Affiliation(s)
- Sylvie Briquet
- Centre d'Immunologie et des Maladies Infectieuses, INSERM, CNRS, Sorbonne Université, Paris, France
| | - Carine Marinach
- Centre d'Immunologie et des Maladies Infectieuses, INSERM, CNRS, Sorbonne Université, Paris, France
| | - Olivier Silvie
- Centre d'Immunologie et des Maladies Infectieuses, INSERM, CNRS, Sorbonne Université, Paris, France
| | - Catherine Vaquero
- Centre d'Immunologie et des Maladies Infectieuses, INSERM, CNRS, Sorbonne Université, Paris, France
| |
Collapse
|
14
|
Fernandes P, Briquet S, Patarot D, Loubens M, Hoareau-Coudert B, Silvie O. The dimerisable Cre recombinase allows conditional genome editing in the mosquito stages of Plasmodium berghei. PLoS One 2020; 15:e0236616. [PMID: 33044964 PMCID: PMC7549836 DOI: 10.1371/journal.pone.0236616] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/01/2020] [Indexed: 01/18/2023] Open
Abstract
Asexual blood stages of the malaria parasite are readily amenable to genetic modification via homologous recombination, allowing functional studies of parasite genes that are not essential in this part of the life cycle. However, conventional reverse genetics cannot be applied for the functional analysis of genes that are essential during asexual blood-stage replication. Various strategies have been developed for conditional mutagenesis of Plasmodium, including recombinase-based gene deletion, regulatable promoters, and mRNA or protein destabilization systems. Among these, the dimerisable Cre (DiCre) recombinase system has emerged as a powerful approach for conditional gene deletion in P. falciparum. In this system, the bacteriophage Cre is expressed in the form of two separate, enzymatically inactive polypeptides, each fused to a different rapamycin-binding protein. Rapamycin-induced heterodimerization of the two components restores recombinase activity. We have implemented the DiCre system in the rodent malaria parasite P. berghei, and show that rapamycin-induced excision of floxed DNA sequences can be achieved with very high efficiency in both mammalian and mosquito parasite stages. This tool can be used to investigate the function of essential genes not only in asexual blood stages, but also in other parts of the malaria parasite life cycle.
Collapse
Affiliation(s)
- Priyanka Fernandes
- Centre d’Immunologie et des Maladies Infectieuses, INSERM, CNRS, CIMI-Paris, Sorbonne Université, Paris, France
| | - Sylvie Briquet
- Centre d’Immunologie et des Maladies Infectieuses, INSERM, CNRS, CIMI-Paris, Sorbonne Université, Paris, France
| | - Delphine Patarot
- Centre d’Immunologie et des Maladies Infectieuses, INSERM, CNRS, CIMI-Paris, Sorbonne Université, Paris, France
| | - Manon Loubens
- Centre d’Immunologie et des Maladies Infectieuses, INSERM, CNRS, CIMI-Paris, Sorbonne Université, Paris, France
| | - Bénédicte Hoareau-Coudert
- UMS PASS, Plateforme de Cytométrie de la Pitié-Salpêtrière (CyPS), Sorbonne Université, Paris, France
| | - Olivier Silvie
- Centre d’Immunologie et des Maladies Infectieuses, INSERM, CNRS, CIMI-Paris, Sorbonne Université, Paris, France
- * E-mail:
| |
Collapse
|
15
|
Putrianti ED, Schmidt-Christensen A, Heussler V, Matuschewski K, Ingmundson A. A Plasmodium cysteine protease required for efficient transition from the liver infection stage. PLoS Pathog 2020; 16:e1008891. [PMID: 32956401 PMCID: PMC7529260 DOI: 10.1371/journal.ppat.1008891] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/01/2020] [Accepted: 08/15/2020] [Indexed: 01/23/2023] Open
Abstract
The transitions between developmental stages are critical points in the Plasmodium life cycle. The development of Plasmodium in the livers of their mammalian hosts bridges malaria transmission and the onset of clinical symptoms elicited by red blood cell infection. The egress of Plasmodium parasites from the liver must be a carefully orchestrated process to ensure a successful switch to the blood stage of infection. Cysteine protease activity is known to be required for liver-stage Plasmodium egress, but the crucial cysteine protease(s) remained unidentified. Here, we characterize a member of the papain-like cysteine protease family, Plasmodium berghei serine repeat antigen 4 (PbSERA4), that is required for efficient initiation of blood-stage infection. Through the generation PbSERA4-specific antisera and the creation of transgenic parasites expressing fluorescently tagged protein, we show that PbSERA4 is expressed and proteolytically processed in the liver and blood stages of infection. Targeted disruption of PbSERA4 results in viable and virulent blood-stage parasites. However, upon transmission from mosquitoes to mice, Pbsera4(-) parasites displayed a reduced capacity to initiate a new round of asexual blood-stage replication. Our results from cultured cells indicate that this defect results from an inability of the PbSERA4-deficient parasites to egress efficiently from infected cells at the culmination of liver-stage development. Protection against infection with wildtype P. berghei could be generated in animals in which Pbsera4(-) parasites failed to establish infection. Our findings confirm that liver-stage merozoite release is an active process and demonstrate that this parasite-encoded cysteine protease contributes to parasite escape from the liver. Plasmodium parasites cause over 200 million cases of malaria every year. When parasites are transmitted by mosquito bite, they initially colonize the liver before they move into the blood and cause disease. During successful transition from the liver into the blood, Plasmodium cloak themselves in host plasma membrane as they egress from the liver cells. Although some aspects of how Plasmodium exit their host hepatocytes appear unique, certain attributes are shared across diverse pathogens. For example, protease activity is required not only for multiple stages of Plasmodium exit, but is also involved in the egress of some bacteria and other protozoan. Here we characterize a protease in Plasmodium berghei that is expressed in the liver and conserved across Plasmodium species. Through gene targeting, we found PbSERA4 is required for efficient egress of Plasmodium from the liver. In the absence of this protease the transition between the liver and blood stages of growth is prolonged due to inefficient parasite release from liver cells. These findings provide new insights into the function of a conserved Plasmodium protease and into the process of Plasmodium escape from the liver.
Collapse
Affiliation(s)
- Elyzana Dewi Putrianti
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
- Metabolism of Microbial Pathogens, Robert Koch Institute, Berlin, Germany
| | - Anja Schmidt-Christensen
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Volker Heussler
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Kai Matuschewski
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
- Department of Molecular Parasitology, Institute of Biology, Humboldt University Berlin, Berlin, Germany
| | - Alyssa Ingmundson
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
- Department of Molecular Parasitology, Institute of Biology, Humboldt University Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
16
|
Moreau CA, Quadt KA, Piirainen H, Kumar H, Bhargav SP, Strauss L, Tolia NH, Wade RC, Spatz JP, Kursula I, Frischknecht F. A function of profilin in force generation during malaria parasite motility that is independent of actin binding. J Cell Sci 2020; 134:jcs233775. [PMID: 32034083 DOI: 10.1242/jcs.233775] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 01/06/2020] [Indexed: 01/20/2023] Open
Abstract
During transmission of malaria-causing parasites from mosquito to mammal, Plasmodium sporozoites migrate at high speed within the skin to access the bloodstream and infect the liver. This unusual gliding motility is based on retrograde flow of membrane proteins and highly dynamic actin filaments that provide short tracks for a myosin motor. Using laser tweezers and parasite mutants, we previously suggested that actin filaments form macromolecular complexes with plasma membrane-spanning adhesins to generate force during migration. Mutations in the actin-binding region of profilin, a near ubiquitous actin-binding protein, revealed that loss of actin binding also correlates with loss of force production and motility. Here, we show that different mutations in profilin, that do not affect actin binding in vitro, still generate lower force during Plasmodium sporozoite migration. Lower force generation inversely correlates with increased retrograde flow suggesting that, like in mammalian cells, the slow down of flow to generate force is the key underlying principle governing Plasmodium gliding motility.
Collapse
Affiliation(s)
- Catherine A Moreau
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Katharina A Quadt
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
- Department of Cellular Biophysics, Max Planck Institute for Medical Research and Laboratory of Biophysical Chemistry, Heidelberg University, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Henni Piirainen
- Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 7, 90220 Oulu, Finland
| | - Hirdesh Kumar
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO 63110, USA
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance and Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Saligram P Bhargav
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
- Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 7, 90220 Oulu, Finland
| | - Léanne Strauss
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Niraj H Tolia
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Rebecca C Wade
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance and Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Heidelberg Institute for Theoretical Studies (HITS), Schloss-Wolfsbrunnenweg 35, 69118 Heidelberg, Germany
| | - Joachim P Spatz
- Department of Cellular Biophysics, Max Planck Institute for Medical Research and Laboratory of Biophysical Chemistry, Heidelberg University, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Inari Kursula
- Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 7, 90220 Oulu, Finland
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| |
Collapse
|
17
|
Affiliation(s)
- Kelly T. Rios
- Department of Biochemistry and Molecular Biology, The Huck Center for Malaria Research, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Scott E. Lindner
- Department of Biochemistry and Molecular Biology, The Huck Center for Malaria Research, Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
18
|
Bennink S, Pradel G. The molecular machinery of translational control in malaria parasites. Mol Microbiol 2019; 112:1658-1673. [PMID: 31531994 DOI: 10.1111/mmi.14388] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2019] [Indexed: 12/30/2022]
Abstract
Translational control regulates the levels of protein synthesized from its transcript and is key for the rapid adjustment of gene expression in response to environmental stimuli. The regulation of translation is of special importance for malaria parasites, which pass through a complex life cycle that includes various replication phases in the different organs of the human and mosquito hosts and a sexual reproduction phase in the mosquito midgut. In particular, the quiescent transmission stages rely on translational control to rapidly adapt to the new environment, once they switch over from the human to the mosquito and vice versa. Three control mechanisms are currently proposed in Plasmodium, (1) global regulation that acts on the translation initiation complex; (2) mRNA-specific regulation, involving cis control elements, mRNA-binding proteins and translational repressors; and (3) induced mRNA decay by the Ccr4-Not and the RNA exosome complex. The main molecules controlling translation are highly conserved in malaria parasites and an increasing number of studies shed light on the interwoven pathways leading to the up or downregulation of protein synthesis in the diverse plasmodial stages. We here highlight recent findings on translational control during life cycle progression of Plasmodium and discuss the molecules involved in regulating protein synthesis.
Collapse
Affiliation(s)
- Sandra Bennink
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Worringerweg 1, 52074, Aachen, Germany
| | - Gabriele Pradel
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Worringerweg 1, 52074, Aachen, Germany
| |
Collapse
|
19
|
Wolanin K, Fontinha D, Sanches-Vaz M, Nyboer B, Heiss K, Mueller AK, Prudêncio M. A crucial role for the C-terminal domain of exported protein 1 during the mosquito and hepatic stages of the Plasmodium berghei life cycle. Cell Microbiol 2019; 21:e13088. [PMID: 31364224 PMCID: PMC6771729 DOI: 10.1111/cmi.13088] [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: 04/30/2019] [Revised: 07/25/2019] [Accepted: 07/28/2019] [Indexed: 12/19/2022]
Abstract
Intracellular Plasmodium parasites develop inside a parasitophorous vacuole (PV), a specialised compartment enclosed by a membrane (PVM) that contains proteins of both host and parasite origin. Although exported protein 1 (EXP1) is one of the earliest described parasitic PVM proteins, its function throughout the Plasmodium life cycle remains insufficiently understood. Here, we show that whereas the N-terminus of Plasmodium berghei EXP1 (PbEXP1) is essential for parasite survival in the blood, parasites lacking PbEXP1's entire C-terminal (CT) domain replicate normally in the blood but cause less severe pathology than their wild-type counterparts. Moreover, truncation of PbEXP1's CT domain not only impairs parasite development in the mosquito but also abrogates PbEXP1 localization to the PVM of intrahepatic parasites, severely limiting their replication and preventing their egress into the blood. Our findings highlight the importance of EXP1 during the Plasmodium life cycle and identify this protein as a promising target for antiplasmodial intervention.
Collapse
Affiliation(s)
- Kamil Wolanin
- Parasitology Unit, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Diana Fontinha
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Margarida Sanches-Vaz
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Britta Nyboer
- Parasitology Unit, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Kirsten Heiss
- Parasitology Unit, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany.,German Centre for Infection Research, Heidelberg Division, Heidelberg, Germany.,PEPperPRINT GmbH, Research & Development Division, Heidelberg, Germany
| | - Ann-Kristin Mueller
- Parasitology Unit, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany.,German Centre for Infection Research, Heidelberg Division, Heidelberg, Germany
| | - Miguel Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
20
|
ApiAP2 Transcription Factors in Apicomplexan Parasites. Pathogens 2019; 8:pathogens8020047. [PMID: 30959972 PMCID: PMC6631176 DOI: 10.3390/pathogens8020047] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/26/2019] [Accepted: 03/28/2019] [Indexed: 12/26/2022] Open
Abstract
Apicomplexan parasites are protozoan organisms that are characterised by complex life cycles and they include medically important species, such as the malaria parasite Plasmodium and the causative agents of toxoplasmosis (Toxoplasma gondii) and cryptosporidiosis (Cryptosporidium spp.). Apicomplexan parasites can infect one or more hosts, in which they differentiate into several morphologically and metabolically distinct life cycle stages. These developmental transitions rely on changes in gene expression. In the last few years, the important roles of different members of the ApiAP2 transcription factor family in regulating life cycle transitions and other aspects of parasite biology have become apparent. Here, we review recent progress in our understanding of the different members of the ApiAP2 transcription factor family in apicomplexan parasites.
Collapse
|
21
|
Costa G, Gildenhard M, Eldering M, Lindquist RL, Hauser AE, Sauerwein R, Goosmann C, Brinkmann V, Carrillo-Bustamante P, Levashina EA. Non-competitive resource exploitation within mosquito shapes within-host malaria infectivity and virulence. Nat Commun 2018; 9:3474. [PMID: 30150763 PMCID: PMC6110728 DOI: 10.1038/s41467-018-05893-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 08/01/2018] [Indexed: 11/22/2022] Open
Abstract
Malaria is a fatal human parasitic disease transmitted by a mosquito vector. Although the evolution of within-host malaria virulence has been the focus of many theoretical and empirical studies, the vector’s contribution to this process is not well understood. Here, we explore how within-vector resource exploitation would impact the evolution of within-host Plasmodium virulence. By combining within-vector dynamics and malaria epidemiology, we develop a mathematical model, which predicts that non-competitive parasitic resource exploitation within-vector restricts within-host parasite virulence. To validate our model, we experimentally manipulate mosquito lipid trafficking and gauge within-vector parasite development and within-host infectivity and virulence. We find that mosquito-derived lipids determine within-host parasite virulence by shaping development (quantity) and metabolic activity (quality) of transmissible sporozoites. Our findings uncover the potential impact of within-vector environment and vector control strategies on the evolution of malaria virulence. The evolution of within-host malaria virulence has been studied, but the vector’s contribution isn’t well understood. Here, Costa et al. show that non-competitive parasitic resource exploitation within-vector, in particular lipid trafficking, restricts within-host infectivity and virulence of the parasite.
Collapse
Affiliation(s)
- G Costa
- Vector Biology Unit, Max Planck Institute for Infection Biology (MPIIB), 10117, Berlin, Germany
| | - M Gildenhard
- Vector Biology Unit, Max Planck Institute for Infection Biology (MPIIB), 10117, Berlin, Germany
| | - M Eldering
- Vector Biology Unit, Max Planck Institute for Infection Biology (MPIIB), 10117, Berlin, Germany.,Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - R L Lindquist
- Immunodynamics, German Rheumatism Research Centre (DRFZ), 10117, Berlin, Germany
| | - A E Hauser
- Immunodynamics, German Rheumatism Research Centre (DRFZ), 10117, Berlin, Germany.,Immune Dynamics and Intravital Microscopy, Charité-Universitätsmedizin, 10117, Berlin, Germany
| | - R Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - C Goosmann
- Microscopy Core Facility, Max Planck Institute for Infection Biology (MPIIB), 10117, Berlin, Germany
| | - V Brinkmann
- Microscopy Core Facility, Max Planck Institute for Infection Biology (MPIIB), 10117, Berlin, Germany
| | - P Carrillo-Bustamante
- Vector Biology Unit, Max Planck Institute for Infection Biology (MPIIB), 10117, Berlin, Germany
| | - E A Levashina
- Vector Biology Unit, Max Planck Institute for Infection Biology (MPIIB), 10117, Berlin, Germany.
| |
Collapse
|
22
|
Cowell AN, Valdivia HO, Bishop DK, Winzeler EA. Exploration of Plasmodium vivax transmission dynamics and recurrent infections in the Peruvian Amazon using whole genome sequencing. Genome Med 2018; 10:52. [PMID: 29973248 PMCID: PMC6032790 DOI: 10.1186/s13073-018-0563-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 06/25/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Plasmodium vivax poses a significant challenge to malaria elimination due to its ability to cause relapsed infections from reactivation of dormant liver parasites called hypnozoites. We analyzed 69 P. vivax whole genome sequences obtained from subjects residing in three different villages along the Peruvian Amazon. This included 23 paired P. vivax samples from subjects who experienced recurrent P. vivax parasitemia following observed treatment with chloroquine and primaquine. METHODS Genomic DNA was extracted from whole blood samples collected from subjects. P. vivax DNA was enriched using selective whole genome amplification and whole genome sequencing. We used single nucleotide polymorphisms (SNPs) from the core P. vivax genome to determine characteristics of the parasite population using discriminant analysis of principal components, maximum likelihood estimation of individual ancestries, and phylogenetic analysis. We estimated the relatedness of the paired samples by calculating the number of segregating sites and using a hidden Markov model approach to estimate identity by descent. RESULTS We present a comprehensive dataset of population genetics of Plasmodium vivax in the Peruvian Amazonian. We define the parasite population structure in this region and demonstrate a novel method for distinguishing homologous relapses from reinfections or heterologous relapses with improved accuracy. The parasite population in this area was quite diverse with an estimated five subpopulations and evidence of a highly heterogeneous ancestry of some of the isolates, similar to previous analyses of P. vivax in this region. Pairwise comparison of recurrent infections determined that there were 12 homologous relapses and 3 likely heterologous relapses with highly related parasites. To the best of our knowledge, this is the first large-scale study to evaluate recurrent P. vivax infections using whole genome sequencing. CONCLUSIONS Whole genome sequencing is a high-resolution tool that can identify P. vivax homologous relapses with increased sensitivity, while also providing data about drug resistance and parasite population genetics. This information is important for evaluating the efficacy of known and novel antirelapse medications in endemic areas and thus advancing the campaign to eliminate malaria.
Collapse
Affiliation(s)
- Annie N Cowell
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA, 92093, USA.
| | - Hugo O Valdivia
- U.S. Naval Medical Research No. 6, Venezuela Ave, Block 36, Bellavista, Callao, Peru
| | - Danett K Bishop
- U.S. Naval Medical Research No. 6, Venezuela Ave, Block 36, Bellavista, Callao, Peru
| | - Elizabeth A Winzeler
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, UC San Diego, 9500 Gilman Dr., La Jolla, CA, 92093, USA
| |
Collapse
|
23
|
Andreae CA, Sessions RB, Virji M, Hill DJ. Bioinformatic analysis of meningococcal Msf and Opc to inform vaccine antigen design. PLoS One 2018; 13:e0193940. [PMID: 29547646 PMCID: PMC5856348 DOI: 10.1371/journal.pone.0193940] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/21/2018] [Indexed: 11/19/2022] Open
Abstract
Neisseria meningitidis is an antigenically and genetically variable Gram-negative bacterium and a causative agent of meningococcal meningitis and septicaemia. Meningococci encode many outer membrane proteins, including Opa, Opc, Msf, fHbp and NadA, identified as being involved in colonisation of the host and evasion of the immune response. Although vaccines are available for the prevention of some types of meningococcal disease, none currently offer universal protection. We have used sequences within the Neisseria PubMLST database to determine the variability of msf and opc in 6,500 isolates. In-silico analysis revealed that although opc is highly conserved, it is not present in all isolates, with most isolates in clonal complex ST-11 lacking a functional opc. In comparison, msf is found in all meningococcal isolates, and displays diversity in the N-terminal domain. We identified 20 distinct Msf sequence variants (Msf SV), associated with differences in number of residues within the putative Vn binding motifs. Moreover, we showed distinct correlations with certain Msf SVs and isolates associated with either hyperinvasive lineages or those clonal complexes associated with a carriage state. We have demonstrated differences in Vn binding between three Msf SVs and generated a cross reactive Msf polyclonal antibody. Our study has highlighted the importance of using large datasets to inform vaccine development and provide further information on the antigenic diversity exhibited by N. meningitidis.
Collapse
Affiliation(s)
- Clio A. Andreae
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | | | - Mumtaz Virji
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Darryl. J. Hill
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
- * E-mail:
| |
Collapse
|
24
|
Kefi M, Mavridis K, Simões ML, Dimopoulos G, Siden-Kiamos I, Vontas J. New rapid one-step PCR diagnostic assay for Plasmodium falciparum infective mosquitoes. Sci Rep 2018; 8:1462. [PMID: 29362379 PMCID: PMC5780459 DOI: 10.1038/s41598-018-19780-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/27/2017] [Indexed: 12/19/2022] Open
Abstract
An essential component of malaria vector control programmes is the detection of Plasmodium falciparum within its mosquito vectors, particularly in the salivary glands where the infective sporozoites reside. Several protocols have been developed for this purpose; however they require dissection of mosquito specimens prior to analysis. Here, a novel one-step RT-qPCR TaqMan diagnostic assay was developed for mosquitoes with infective Plasmodium falciparum sporozoites in the salivary glands. It is based on detection of the sporozoite-specific Pfslarp and Pfplp1 gene transcripts. These transcripts were chosen based on bioinformatics analysis, and experimentally verified to be overexpressed in the salivary gland sporozoite stage of the parasite compared to other mosquito parasite stages. The proof of principle and the performance of the assay were demonstrated using RNAlater preserved mosquito samples. Tests of analytical sensitivity showed the novel TaqMan assay to be 100% accurate, although its performance in the field needs to be further demonstrated. This method has no requirement for dissection and post-PCR processing and thus is simple and rapid to perform in individual mosquitoes or mosquito pools. It can be used in single or multiplex formats also targeting additional markers expressed in different tissues, such as detoxification enzymes associated with insecticide resistance.
Collapse
Affiliation(s)
- Mary Kefi
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 70013, Greece.,Department of Biology, University of Crete, VassilikaVouton, Heraklion, 70013, Greece
| | - Konstantinos Mavridis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 70013, Greece
| | - Maria L Simões
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615N Wolfe St, Baltimore, MD, 21205, USA
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615N Wolfe St, Baltimore, MD, 21205, USA
| | - Inga Siden-Kiamos
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 70013, Greece.
| | - John Vontas
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, 70013, Greece.,Pesticide Science Laboratory, Department of Crop Science, Agricultural University of Athens, 11855, Athens, Greece
| |
Collapse
|
25
|
Nash SD, Prevots DR, Kabyemela E, Khasa YP, Lee KL, Fried M, Duffy PE. A Malaria-Resistant Phenotype with Immunological Correlates in a Tanzanian Birth Cohort Exposed to Intense Malaria Transmission. Am J Trop Med Hyg 2017; 96:1190-1196. [PMID: 28500801 DOI: 10.4269/ajtmh.16-0554] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
AbstractMalaria incidence is highly heterogeneous even in areas of high transmission, although no conclusive evidence exists that innate or naturally acquired resistance can prevent infection over an extended period of time. This longitudinal study examined immunoparasitological evidence for a malaria-resistant phenotype in which children do not develop malaria despite an extended period of exposure to parasites. Within a birth cohort followed from 2002 to 2006 in Muheza, Tanzania, an area of intense transmission, children (N = 687) provided blood smears biweekly during infancy and monthly thereafter. Maternal and childhood characteristics were obtained, cord-blood cytokines were measured, and antibody responses were assayed as measures of stage-specific exposure. Sixty-three (9.2%) children had no blood smear-positive slides over 2 years of follow-up (range: 1-3.5 years) and were identified as malaria resistant. Malaria-resistant children were similar to other children with respect to completeness of follow-up and all maternal and childhood characteristics except residence area. Antibody seroprevalence was similar for two sporozoite antigens, but malaria-resistant children had a lower antibody seroprevalence to merozoite antigens merozoite surface protein 1 (5.4% versus 30.2%; P < 0.0001) and apical membrane antigen 1 (7.2% versus 33.3%; P < 0.0001). Malaria-resistant children had higher cytokine levels in cord blood, particularly interleukin-1β. In summary, a subset of children living in an area of intense transmission was exposed to malaria parasites, but never developed patent parasitemia; this phenotype was associated with a distinct cytokine profile at birth and antibody profile during infancy. Further research with malaria-resistant children may identify mechanisms for naturally acquired immunity.
Collapse
Affiliation(s)
- Scott D Nash
- Epidemiology Unit, Laboratory of Clinical Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland.,Laboratory of Malaria Immunology and Vaccinology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland
| | - D Rebecca Prevots
- Epidemiology Unit, Laboratory of Clinical Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | | | - Yogender P Khasa
- Department of Microbiology, University of Delhi South Campus, New Delhi, India
| | - Kun-Lin Lee
- Laboratory of Malaria Immunology and Vaccinology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland
| | - Michal Fried
- Laboratory of Malaria Immunology and Vaccinology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland
| |
Collapse
|
26
|
Cubi R, Vembar SS, Biton A, Franetich J, Bordessoulles M, Sossau D, Zanghi G, Bosson‐Vanga H, Benard M, Moreno A, Dereuddre‐Bosquet N, Le Grand R, Scherf A, Mazier D. Laser capture microdissection enables transcriptomic analysis of dividing and quiescent liver stages of Plasmodium relapsing species. Cell Microbiol 2017; 19:e12735. [PMID: 28256794 PMCID: PMC5516136 DOI: 10.1111/cmi.12735] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 02/24/2017] [Indexed: 01/15/2023]
Abstract
Dormant liver stage forms (hypnozoites) of the malaria parasite Plasmodium vivax present major hurdles to control and eradicate infection. Despite major research efforts, the molecular composition of hypnozoites remains ill defined. Here, we applied a combination of state-of-the-art technologies to generate the first transcriptome of hypnozoites. We developed a robust laser dissection microscopy protocol to isolate individual Plasmodium cynomolgi hypnozoites and schizonts from infected monkey hepatocytes and optimized RNA-seq analysis to obtain the first transcriptomes of these stages. Comparative transcriptomic analysis identified 120 transcripts as being differentially expressed in the hypnozoite stage relative to the dividing liver schizont, with 69 and 51 mRNAs being up- or down-regulated, respectively, in the hypnozoites. This lead to the identification of potential markers of commitment to and maintenance of the dormant state of the hypnozoite including three transcriptional regulators of the ApiAP2 family, one of which is unique to P. cynomolgi and P. vivax, and the global translational repressor, eIF2a kinase eIK2, all of which are upregulated in the hypnozoite. Together, this work not only provides a primary experimentally-derived list of molecular markers of hypnozoites but also identifies transcriptional and posttranscriptional regulation of gene expression as potentially being key to establishing and maintaining quiescence.
Collapse
Affiliation(s)
- Roger Cubi
- Centre d'Immunologie et des Maladies Infectieuses, CNRS ERL8255, INSERM U1135Sorbonne Universités, UPMC Univ Paris 06ParisFrance
| | - Shruthi S. Vembar
- Unité Biologie des Interactions Hôte‐Parasite—Institut PasteurParisFrance
- CNRS ERL 9195ParisFrance
- INSERM U1201ParisFrance
| | - Anne Biton
- Centre de BioinformatiqueBiostatistique et Biologie Intégrative (C3BI, USR 3756 Institut Pasteur et CNRS)ParisFrance
| | - Jean‐Francois Franetich
- Centre d'Immunologie et des Maladies Infectieuses, CNRS ERL8255, INSERM U1135Sorbonne Universités, UPMC Univ Paris 06ParisFrance
| | - Mallaury Bordessoulles
- Centre d'Immunologie et des Maladies Infectieuses, CNRS ERL8255, INSERM U1135Sorbonne Universités, UPMC Univ Paris 06ParisFrance
| | - Daniel Sossau
- Centre d'Immunologie et des Maladies Infectieuses, CNRS ERL8255, INSERM U1135Sorbonne Universités, UPMC Univ Paris 06ParisFrance
- Department of DermatologyEberhard Karls UniversityTübingenGermany
| | - Gigliola Zanghi
- Centre d'Immunologie et des Maladies Infectieuses, CNRS ERL8255, INSERM U1135Sorbonne Universités, UPMC Univ Paris 06ParisFrance
| | - Henriette Bosson‐Vanga
- Centre d'Immunologie et des Maladies Infectieuses, CNRS ERL8255, INSERM U1135Sorbonne Universités, UPMC Univ Paris 06ParisFrance
| | | | - Alicia Moreno
- AP‐HP, Hôpital St. AntoineService de Parasitologie‐Mycologie75012ParisFrance
| | - Nathalie Dereuddre‐Bosquet
- Immunology of Viral Infections and Autoimmune DiseasesCEA—Université Paris Sud 1—INSERM U1184Fontenay‐aux‐RosesFrance
| | - Roger Le Grand
- Immunology of Viral Infections and Autoimmune DiseasesCEA—Université Paris Sud 1—INSERM U1184Fontenay‐aux‐RosesFrance
| | - Artur Scherf
- Unité Biologie des Interactions Hôte‐Parasite—Institut PasteurParisFrance
- CNRS ERL 9195ParisFrance
- INSERM U1201ParisFrance
| | - Dominique Mazier
- Centre d'Immunologie et des Maladies Infectieuses, CNRS ERL8255, INSERM U1135Sorbonne Universités, UPMC Univ Paris 06ParisFrance
- AP‐HP, Groupe Hospitalier Pitié‐Salpêtrière, Service Parasitologie‐MycologieParisFrance
| |
Collapse
|
27
|
Vaughan AM, Kappe SHI. Malaria Parasite Liver Infection and Exoerythrocytic Biology. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a025486. [PMID: 28242785 DOI: 10.1101/cshperspect.a025486] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
In their infection cycle, malaria parasites undergo replication and population expansions within the vertebrate host and the mosquito vector. Host infection initiates with sporozoite invasion of hepatocytes, followed by a dramatic parasite amplification event during liver stage parasite growth and replication within hepatocytes. Each liver stage forms up to 90,000 exoerythrocytic merozoites, which are in turn capable of initiating a blood stage infection. Liver stages not only exploit host hepatocyte resources for nutritional needs but also endeavor to prevent hepatocyte cell death and detection by the host's immune system. Research over the past decade has identified numerous parasite factors that play a critical role during liver infection and has started to delineate a complex web of parasite-host interactions that sustain successful parasite colonization of the mammalian host. Targeting the parasites' obligatory infection of the liver as a gateway to the blood, with drugs and vaccines, constitutes the most effective strategy for malaria eradication, as it would prevent clinical disease and onward transmission of the parasite.
Collapse
Affiliation(s)
- Ashley M Vaughan
- Center for Infectious Disease Research, formerly Seattle Biomedical Research Institute, Seattle, Washington 98109
| | - Stefan H I Kappe
- Center for Infectious Disease Research, formerly Seattle Biomedical Research Institute, Seattle, Washington 98109.,Department of Global Health, University of Washington, Seattle, Washington 98195
| |
Collapse
|
28
|
Kreutzfeld O, Müller K, Matuschewski K. Engineering of Genetically Arrested Parasites (GAPs) For a Precision Malaria Vaccine. Front Cell Infect Microbiol 2017; 7:198. [PMID: 28620583 PMCID: PMC5450620 DOI: 10.3389/fcimb.2017.00198] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 05/04/2017] [Indexed: 01/08/2023] Open
Abstract
Continuous stage conversion and swift changes in the antigenic repertoire in response to acquired immunity are hallmarks of complex eukaryotic pathogens, including Plasmodium species, the causative agents of malaria. Efficient elimination of Plasmodium liver stages prior to blood infection is one of the most promising malaria vaccine strategies. Here, we describe different genetically arrested parasites (GAPs) that have been engineered in Plasmodium berghei, P. yoelii and P. falciparum and compare their vaccine potential. A better understanding of the immunological mechanisms of prime and boost by arrested sporozoites and experimental strategies to enhance vaccine efficacy by further engineering existing GAPs into a more immunogenic form hold promise for continuous improvements of GAP-based vaccines. A critical hurdle for vaccines that elicit long-lasting protection against malaria, such as GAPs, is safety and efficacy in vulnerable populations. Vaccine research should focus on solutions toward turning malaria into a vaccine-preventable disease, which would offer an exciting new path of malaria control.
Collapse
Affiliation(s)
- Oriana Kreutzfeld
- Department of Molecular Parasitology, Institute of Biology, Humboldt UniversityBerlin, Germany
| | - Katja Müller
- Department of Molecular Parasitology, Institute of Biology, Humboldt UniversityBerlin, Germany
| | - Kai Matuschewski
- Department of Molecular Parasitology, Institute of Biology, Humboldt UniversityBerlin, Germany
| |
Collapse
|
29
|
Moreau CA, Bhargav SP, Kumar H, Quadt KA, Piirainen H, Strauss L, Kehrer J, Streichfuss M, Spatz JP, Wade RC, Kursula I, Frischknecht F. A unique profilin-actin interface is important for malaria parasite motility. PLoS Pathog 2017; 13:e1006412. [PMID: 28552953 PMCID: PMC5464670 DOI: 10.1371/journal.ppat.1006412] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 06/08/2017] [Accepted: 05/16/2017] [Indexed: 11/30/2022] Open
Abstract
Profilin is an actin monomer binding protein that provides ATP-actin for incorporation into actin filaments. In contrast to higher eukaryotic cells with their large filamentous actin structures, apicomplexan parasites typically contain only short and highly dynamic microfilaments. In apicomplexans, profilin appears to be the main monomer-sequestering protein. Compared to classical profilins, apicomplexan profilins contain an additional arm-like β-hairpin motif, which we show here to be critically involved in actin binding. Through comparative analysis using two profilin mutants, we reveal this motif to be implicated in gliding motility of Plasmodium berghei sporozoites, the rapidly migrating forms of a rodent malaria parasite transmitted by mosquitoes. Force measurements on migrating sporozoites and molecular dynamics simulations indicate that the interaction between actin and profilin fine-tunes gliding motility. Our data suggest that evolutionary pressure to achieve efficient high-speed gliding has resulted in a unique profilin-actin interface in these parasites. The malaria parasite Plasmodium has two invasive forms that migrate across different tissue barriers, the ookinete and the very rapidly migrating sporozoite. Previous work has shown that the motility of these and related parasites (e.g. Toxoplasma gondii) depends on a highly dynamic actin cytoskeleton and retrograde flow of surface adhesins. These unusual actin dynamics are due to the divergent structure of protozoan actins and the actions of actin-binding proteins, which can have non-canonical functions in these parasites. Profilin is one of the most important and most investigated actin-binding proteins, which binds ADP-actin and catalyzes ADP-ATP exchange to then promote actin polymerization. Parasite profilins bind monomeric actin and contain an additional domain compared to canonical profilins. Here we show that this additional domain of profilin is critical for actin binding and rapid sporozoite motility but has little impact on the slower ookinete. Sporozoites of a parasite line carrying mutations in this domain cannot translate force production and retrograde flow into optimal parasite motility. Using molecular dynamics simulations, we find that differences between mutant parasites in their capacity to migrate can be traced back to a single hydrogen bond at the actin-profilin interface.
Collapse
Affiliation(s)
- Catherine A. Moreau
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Saligram P. Bhargav
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
- Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Hirdesh Kumar
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), Heidelberg, Germany
| | - Katharina A. Quadt
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
- Institute for Physical Chemistry, Biophysical Chemistry, Heidelberg University, Heidelberg, Germany
| | - Henni Piirainen
- Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Léanne Strauss
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Jessica Kehrer
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Martin Streichfuss
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
- Institute for Physical Chemistry, Biophysical Chemistry, Heidelberg University, Heidelberg, Germany
| | - Joachim P. Spatz
- Institute for Physical Chemistry, Biophysical Chemistry, Heidelberg University, Heidelberg, Germany
- Department of Cellular Biophysics, Max-Planck Institute for Medical Research, Heidelberg, Germany
| | - Rebecca C. Wade
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), Heidelberg, Germany
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance and Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Heidelberg, Germany
| | - Inari Kursula
- Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
- Department of Biomedicine, University of Bergen, Bergen, Norway
- * E-mail: (IK); (FF)
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
- * E-mail: (IK); (FF)
| |
Collapse
|
30
|
Petersen W, Stenzel W, Silvie O, Blanz J, Saftig P, Matuschewski K, Ingmundson A. Sequestration of cholesterol within the host late endocytic pathway restricts liver-stage Plasmodium development. Mol Biol Cell 2017; 28:726-735. [PMID: 28122820 PMCID: PMC5349780 DOI: 10.1091/mbc.e16-07-0531] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 01/18/2017] [Accepted: 01/18/2017] [Indexed: 11/17/2022] Open
Abstract
While lysosomes are degradative compartments and one of the defenses against invading pathogens, they are also hubs of metabolic activity. Late endocytic compartments accumulate around Plasmodium berghei liver-stage parasites during development, and whether this is a host defense strategy or active recruitment by the parasites is unknown. In support of the latter hypothesis, we observed that the recruitment of host late endosomes (LEs) and lysosomes is reduced in uis4- parasites, which lack a parasitophorous vacuole membrane protein and arrest during liver-stage development. Analysis of parasite development in host cells deficient for late endosomal or lysosomal proteins revealed that the Niemann-Pick type C (NPC) proteins, which are involved in cholesterol export from LEs, and the lysosome-associated membrane proteins (LAMP) 1 and 2 are important for robust liver-stage P. berghei growth. Using the compound U18666A, which leads to cholesterol sequestration in LEs similar to that seen in NPC- and LAMP-deficient cells, we show that the restriction of parasite growth depends on cholesterol sequestration and that targeting this process can reduce parasite burden in vivo. Taken together, these data reveal that proper LE and lysosome function positively contributes to liver-stage Plasmodium development.
Collapse
Affiliation(s)
- Wiebke Petersen
- Molecular Parasitology, Humboldt University, 10115 Berlin, Germany
- Max Planck Institute for Infection Biology, 10117 Berlin, Germany
| | - Werner Stenzel
- Institute for Neuropathology, Charité-Universitätsmedizin, 10117 Berlin, Germany
| | - Olivier Silvie
- Institut National de la Santé et de la Recherche Médicale, U1135, Centre d'Immunologie et des Maladies Infectieuses, F-75013 Paris, France
| | - Judith Blanz
- Institute of Biochemistry, Christian-Albrechts-University of Kiel, 24098 Kiel, Germany
| | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts-University of Kiel, 24098 Kiel, Germany
| | - Kai Matuschewski
- Molecular Parasitology, Humboldt University, 10115 Berlin, Germany
- Max Planck Institute for Infection Biology, 10117 Berlin, Germany
| | - Alyssa Ingmundson
- Molecular Parasitology, Humboldt University, 10115 Berlin, Germany
- Max Planck Institute for Infection Biology, 10117 Berlin, Germany
| |
Collapse
|
31
|
Plasmodium berghei EXP-1 interacts with host Apolipoprotein H during Plasmodium liver-stage development. Proc Natl Acad Sci U S A 2017; 114:E1138-E1147. [PMID: 28137845 DOI: 10.1073/pnas.1606419114] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The first, obligatory replication phase of malaria parasite infections is characterized by rapid expansion and differentiation of single parasites in liver cells, resulting in the formation and release of thousands of invasive merozoites into the bloodstream. Hepatic Plasmodium development occurs inside a specialized membranous compartment termed the parasitophorous vacuole (PV). Here, we show that, during the parasite's hepatic replication, the C-terminal region of the parasitic PV membrane protein exported protein 1 (EXP-1) binds to host Apolipoprotein H (ApoH) and that this molecular interaction plays a pivotal role for successful Plasmodium liver-stage development. Expression of a truncated EXP-1 protein, missing the specific ApoH interaction site, or down-regulation of ApoH expression in either hepatic cells or mouse livers by RNA interference resulted in impaired intrahepatic development. Furthermore, infection of mice with sporozoites expressing a truncated version of EXP-1 resulted in both a significant reduction of liver burden and delayed blood-stage patency, leading to a disease outcome different from that generally induced by infection with wild-type parasites. This study identifies a host-parasite protein interaction during the hepatic stage of infection by Plasmodium parasites. The identification of such vital interactions may hold potential toward the development of novel malaria prevention strategies.
Collapse
|
32
|
Koussis K, Goulielmaki E, Chalari A, Withers-Martinez C, Siden-Kiamos I, Matuschewski K, Loukeris TG. Targeted Deletion of a Plasmodium Site-2 Protease Impairs Life Cycle Progression in the Mammalian Host. PLoS One 2017; 12:e0170260. [PMID: 28107409 PMCID: PMC5249076 DOI: 10.1371/journal.pone.0170260] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/01/2017] [Indexed: 12/26/2022] Open
Abstract
Site-2 proteases (S2P) belong to the M50 family of metalloproteases, which typically perform essential roles by mediating activation of membrane–bound transcription factors through regulated intramembrane proteolysis (RIP). Protease-dependent liberation of dormant transcription factors triggers diverse cellular responses, such as sterol regulation, Notch signalling and the unfolded protein response. Plasmodium parasites rely on regulated proteolysis for controlling essential pathways throughout the life cycle. In this study we examine the Plasmodium-encoded S2P in a murine malaria model and show that it is expressed in all stages of Plasmodium development. Localisation studies by endogenous gene tagging revealed that in all invasive stages the protein is in close proximity to the nucleus. Ablation of PbS2P by reverse genetics leads to reduced growth rates during liver and blood infection and, hence, virulence attenuation. Strikingly, absence of PbS2P was compatible with parasite life cycle progression in the mosquito and mammalian hosts under physiological conditions, suggesting redundant or dispensable roles in vivo.
Collapse
Affiliation(s)
- Konstantinos Koussis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
- * E-mail:
| | - Evi Goulielmaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Anna Chalari
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
- Department of Biology, University of Crete, Heraklion, Greece
| | | | - Inga Siden-Kiamos
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Kai Matuschewski
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
- Institute of Biology, Humboldt University, Berlin, Germany
| | - Thanasis G. Loukeris
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| |
Collapse
|
33
|
Al-Nihmi FMA, Kolli SK, Reddy SR, Mastan BS, Togiri J, Maruthi M, Gupta R, Sijwali PS, Mishra S, Kumar KA. A Novel and Conserved Plasmodium Sporozoite Membrane Protein SPELD is Required for Maturation of Exo-erythrocytic Forms. Sci Rep 2017; 7:40407. [PMID: 28067322 PMCID: PMC5220379 DOI: 10.1038/srep40407] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 12/06/2016] [Indexed: 02/07/2023] Open
Abstract
Plasmodium sporozoites are the infective forms of malaria parasite to vertebrate host and undergo dramatic changes in their transcriptional repertoire during maturation in mosquito salivary glands. We report here the role of a novel and conserved Plasmodium berghei protein encoded by PBANKA_091090 in maturation of Exo-erythrocytic Forms (EEFs) and designate it as Sporozoite surface Protein Essential for Liver stage Development (PbSPELD). PBANKA_091090 was previously annotated as PB402615.00.0 and its transcript was recovered at maximal frequency in the Serial Analysis of the Gene Expression (SAGE) of Plasmodium berghei salivary gland sporozoites. An orthologue of this transcript was independently identified in Plasmodium vivax sporozoite microarrays and was designated as Sporozoite Conserved Orthologous Transcript-2 (scot-2). Functional characterization through reverse genetics revealed that PbSPELD is essential for Plasmodium liver stage maturation. mCherry transgenic of PbSPELD localized the protein to plasma membrane of sporozoites and early EEFs. Global microarray analysis of pbspeld ko revealed EEF attenuation being associated with down regulation of genes central to general transcription, cell cycle, proteosome and cadherin signaling. pbspeld mutant EEFs induced pre-erythrocytic immunity with 50% protective efficacy. Our studies have implications for attenuating the human Plasmodium liver stages by targeting SPELD locus.
Collapse
Affiliation(s)
| | - Surendra Kumar Kolli
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Segireddy Rameswara Reddy
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Babu S Mastan
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Jyothi Togiri
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Mulaka Maruthi
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| | - Roshni Gupta
- Division of Parasitology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Puran Singh Sijwali
- CSIR-Centre for Cellular and Molecular Biology, Habsiguda, Uppal Road, Hyderabad 500007, India
| | - Satish Mishra
- Division of Parasitology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Kota Arun Kumar
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India
| |
Collapse
|
34
|
Distinct Prominent Roles for Enzymes of Plasmodium berghei Heme Biosynthesis in Sporozoite and Liver Stage Maturation. Infect Immun 2016; 84:3252-3262. [PMID: 27600503 DOI: 10.1128/iai.00148-16] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 08/29/2016] [Indexed: 11/20/2022] Open
Abstract
Malarial parasites have evolved complex regulation of heme supply and disposal to adjust to heme-rich and -deprived host environments. In addition to its own pathway for heme biosynthesis, Plasmodium likely harbors mechanisms for heme scavenging from host erythrocytes. Elaborate compartmentalization of de novo heme synthesis into three subcellular locations, including the vestigial plastid organelle, indicates critical roles in life cycle progression. In this study, we systematically profile the essentiality of heme biosynthesis by targeted gene deletion of enzymes in early steps of this pathway. We show that disruption of endogenous heme biosynthesis leads to a first detectable defect in oocyst maturation and sporogony in the Anopheles vector, whereas blood stage propagation, colonization of mosquito midguts, or initiation of oocyst development occurs indistinguishably from that of wild-type parasites. Although sporozoites are produced by parasites lacking an intact pathway for heme biosynthesis, they are absent from mosquito salivary glands, indicative of a vital role for heme biosynthesis only in sporozoite maturation. Rescue of the first defect in sporogony permitted analysis of potential roles in liver stages. We show that liver stage parasites benefit from but do not strictly depend upon their own aminolevulinic acid synthase and that they can scavenge aminolevulinic acid from the host environment. Together, our experimental genetics analysis of Plasmodium enzymes for heme biosynthesis exemplifies remarkable shifts between the use of endogenous and host resources during life cycle progression.
Collapse
|
35
|
Speake C, Pichugin A, Sahu T, Malkov V, Morrison R, Pei Y, Juompan L, Milman N, Zarling S, Anderson C, Wong-Madden S, Wendler J, Ishizuka A, MacMillen ZW, Garcia V, Kappe SHI, Krzych U, Duffy PE. Identification of Novel Pre-Erythrocytic Malaria Antigen Candidates for Combination Vaccines with Circumsporozoite Protein. PLoS One 2016; 11:e0159449. [PMID: 27434123 PMCID: PMC4951032 DOI: 10.1371/journal.pone.0159449] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 07/01/2016] [Indexed: 12/17/2022] Open
Abstract
Malaria vaccine development has been hampered by the limited availability of antigens identified through conventional discovery approaches, and improvements are needed to enhance the efficacy of the leading vaccine candidate RTS,S that targets the circumsporozoite protein (CSP) of the infective sporozoite. Here we report a transcriptome-based approach to identify novel pre-erythrocytic vaccine antigens that could potentially be used in combination with CSP. We hypothesized that stage-specific upregulated genes would enrich for protective vaccine targets, and used tiling microarray to identify P. falciparum genes transcribed at higher levels during liver stage versus sporozoite or blood stages of development. We prepared DNA vaccines for 21 genes using the predicted orthologues in P. yoelii and P. berghei and tested their efficacy using different delivery methods against pre-erythrocytic malaria in rodent models. In our primary screen using P. yoelii in BALB/c mice, we found that 16 antigens significantly reduced liver stage parasite burden. In our confirmatory screen using P. berghei in C57Bl/6 mice, we confirmed 6 antigens that were protective in both models. Two antigens, when combined with CSP, provided significantly greater protection than CSP alone in both models. Based on the observations reported here, transcriptional patterns of Plasmodium genes can be useful in identifying novel pre-erythrocytic antigens that induce protective immunity alone or in combination with CSP.
Collapse
MESH Headings
- Animals
- Antibodies, Protozoan/immunology
- Antibodies, Protozoan/therapeutic use
- Antigens, Protozoan/immunology
- Female
- Humans
- Malaria Vaccines/genetics
- Malaria Vaccines/immunology
- Malaria Vaccines/therapeutic use
- Malaria, Falciparum/drug therapy
- Malaria, Falciparum/immunology
- Malaria, Falciparum/parasitology
- Mice
- Mice, Inbred C57BL
- Plasmodium falciparum/immunology
- Plasmodium falciparum/pathogenicity
- Plasmodium yoelii/immunology
- Protozoan Proteins/immunology
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Vaccines, DNA/therapeutic use
Collapse
Affiliation(s)
- Cate Speake
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Alexander Pichugin
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Tejram Sahu
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Vlad Malkov
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Robert Morrison
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ying Pei
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Laure Juompan
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Neta Milman
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Stasya Zarling
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Charles Anderson
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Sharon Wong-Madden
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jason Wendler
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Andrew Ishizuka
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Zachary W. MacMillen
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Valentino Garcia
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Stefan H. I. Kappe
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Urszula Krzych
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
| | - Patrick E. Duffy
- 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
|
36
|
Bane KS, Lepper S, Kehrer J, Sattler JM, Singer M, Reinig M, Klug D, Heiss K, Baum J, Mueller AK, Frischknecht F. The Actin Filament-Binding Protein Coronin Regulates Motility in Plasmodium Sporozoites. PLoS Pathog 2016; 12:e1005710. [PMID: 27409081 PMCID: PMC4943629 DOI: 10.1371/journal.ppat.1005710] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 05/26/2016] [Indexed: 11/21/2022] Open
Abstract
Parasites causing malaria need to migrate in order to penetrate tissue barriers and enter host cells. Here we show that the actin filament-binding protein coronin regulates gliding motility in Plasmodium berghei sporozoites, the highly motile forms of a rodent malaria-causing parasite transmitted by mosquitoes. Parasites lacking coronin show motility defects that impair colonization of the mosquito salivary glands but not migration in the skin, yet result in decreased transmission efficiency. In non-motile sporozoites low calcium concentrations mediate actin-independent coronin localization to the periphery. Engagement of extracellular ligands triggers an intracellular calcium release followed by the actin-dependent relocalization of coronin to the rear and initiation of motility. Mutational analysis and imaging suggest that coronin organizes actin filaments for productive motility. Using coronin-mCherry as a marker for the presence of actin filaments we found that protein kinase A contributes to actin filament disassembly. We finally speculate that calcium and cAMP-mediated signaling regulate a switch from rapid parasite motility to host cell invasion by differentially influencing actin dynamics. Parasites causing malaria are transmitted by mosquitoes and need to migrate to cross tissue barriers. The form of the parasite transmitted by the mosquito, the so-called sporozoite, needs motility to enter the salivary glands, to migrate within the skin and to enter into blood capillaries and eventually hepatocytes, where the parasites differentiate into thousands of merozoites that invade red blood cells. Sporozoite motility is based on an actin-myosin motor, as is the case in many other eukaryotic cells. However, most eukaryotic cells move much slower than sporozoites. How these parasites reach their high speed is not clear but current evidence suggests that actin filaments need to be organized by either actin-binding proteins or membrane proteins that link the filaments to an extracellular substrate. The present study explores the role of the actin filament-binding protein coronin in the motility of sporozoites of the rodent model parasite Plasmodium berghei. We found that the deletion of P. berghei coronin leads to defects in parasite motility and thus lower infection of mosquito salivary glands, which translates into less efficient transmission of the parasites. Our experiments suggest that coronin organizes actin filaments to achieve rapid and directional motility. We also identify two signaling pathways that converge to regulate actin filament dynamics and suggest that they play a role in switching the parasite from its motility mode to a cell invasion mode.
Collapse
Affiliation(s)
- Kartik S. Bane
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | - Simone Lepper
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | - Jessica Kehrer
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | - Julia M. Sattler
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | - Mirko Singer
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | - Miriam Reinig
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | - Dennis Klug
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | - Kirsten Heiss
- Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
- Malva GmbH, Heidelberg, Germany
| | - Jake Baum
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Ann-Kristin Mueller
- Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
37
|
Kersting D, Krüger M, Sattler JM, Mueller AK, Kaiser A. A suggested vital function for eIF-5A and dhs genes during murine malaria blood-stage infection. FEBS Open Bio 2016; 6:860-72. [PMID: 27516964 PMCID: PMC4971841 DOI: 10.1002/2211-5463.12093] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 05/07/2016] [Accepted: 05/29/2016] [Indexed: 12/30/2022] Open
Abstract
The biological function of the post-translational modification hypusine in the eukaryotic initiation factor 5A (EIF-5A) in eukaryotes is still not understood. Hypusine is formed by two sequential enzymatic steps at a specific lysine residue in the precursor protein EIF-5A. One important biological function of EIF-5A which was recently identified is the translation of polyproline-rich mRNA, suggesting its biological relevance in a variety of biological processes. Hypusinated eIF-5A controls the proliferation of cancer cells and inflammatory processes in malaria. It was shown that pharmacological inhibition of the enzymes involved in this pathway, deoxyhypusine synthase (DHS) and the deoxyhypusine hydroxylase (DOHH), arrested the growth of malaria parasites. Down-regulation of both the malarial eIF-5A and dhs genes by in vitro and in vivo silencing led to decreased transcript levels and protein expression and, in turn, to low parasitemia, confirming a critical role of hypusination in eIF-5A for proliferation in Plasmodium. To further investigate whether eIF-5A and the activating enzyme DHS are essential for Plasmodium erythrocytic stages, targeted gene disruption was performed in the rodent malaria parasite Plasmodium berghei. Full disruption of both genes was not successful; instead parasites harboring the episome for eIF-5A and dhs genes were obtained, suggesting that these genes may perform vital functions during the pathogenic blood cell stage. Next, a knock-in strategy was pursued for both endogenous genes eIF-5A and dhs from P. berghei. The latter resulted in viable recombinant parasites, strengthening the observation that they might be essential for proliferation during asexual development of the malaria parasite.
Collapse
Affiliation(s)
- David Kersting
- Institute for Pharmacogenetics Medical Research Centre University Duisburg-Essen Germany
| | - Mirko Krüger
- Institute for Pharmacogenetics Medical Research Centre University Duisburg-Essen Germany
| | - Julia M Sattler
- Parasitology Unit Centre for Infectious Diseases University Hospital Heidelberg Germany; Centre for Infectious Diseases, Integrative Parasitology University Hospital Heidelberg Germany
| | - Ann-Kristin Mueller
- Parasitology Unit Centre for Infectious Diseases University Hospital Heidelberg Germany; German Center for Infectious Diseases Heidelberg Germany
| | - Annette Kaiser
- Institute for Pharmacogenetics Medical Research Centre University Duisburg-Essen Germany
| |
Collapse
|
38
|
van der Velden M, Rijpma SR, Verweij V, van Gemert GJ, Chevalley-Maurel S, van de Vegte-Bolmer M, Franke-Fayard BM, Russel FGM, Janse CJ, Sauerwein RW, Koenderink JB. Protective Efficacy Induced by Genetically Attenuated Mid-to-Late Liver-Stage Arresting Plasmodium berghei Δmrp2 Parasites. Am J Trop Med Hyg 2016; 95:378-82. [PMID: 27296385 DOI: 10.4269/ajtmh.16-0226] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 05/04/2016] [Indexed: 01/05/2023] Open
Abstract
Whole parasite immunization strategies employing genetically attenuated parasites (GAP), which arrest during liver-stage development, have been applied successfully for induction of sterile malaria protection in rodents. Recently, we generated a Plasmodium berghei GAP-lacking expression of multidrug resistance-associated protein (MRP2) (PbΔmrp2) that was capable of partial schizogony in hepatocytes but showed complete growth arrest. Here, we investigated the protective efficacy after intravenous (IV) immunization of BALB/c and C57BL/6J mice with PbΔmrp2 sporozoites. Low-dose immunization using 400 PbΔmrp2 sporozoites induced 100% sterile protection in BALB/c mice after IV challenge with 10,000 wild-type sporozoites. In addition, almost full protection (90%) was obtained after three immunizations with 10,000 sporozoites in C57BL/6J mice. Parasite liver loads in nonprotected PbΔmrp2-challenged C57BL/6J mice were reduced by 86% ± 5% on average compared with naive control mice. The mid-to-late arresting PbΔmrp2 GAP was equipotent in induction of protective immunity to the early arresting PbΔb9Δslarp GAP. The combined data support a clear basis for further exploration of Plasmodium falciparum parasites lacking mrp2 as a suitable GAP vaccine candidate.
Collapse
Affiliation(s)
- Maarten van der Velden
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sanna R Rijpma
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Vivienne Verweij
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Séverine Chevalley-Maurel
- Leiden Malaria Research Group, Department of Parasitology, Center of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Blandine M Franke-Fayard
- Leiden Malaria Research Group, Department of Parasitology, Center of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Frans G M Russel
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Chris J Janse
- Leiden Malaria Research Group, Department of Parasitology, Center of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jan B Koenderink
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
39
|
Protective efficacy and safety of liver stage attenuated malaria parasites. Sci Rep 2016; 6:26824. [PMID: 27241521 PMCID: PMC4886212 DOI: 10.1038/srep26824] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 05/09/2016] [Indexed: 12/15/2022] Open
Abstract
During the clinically silent liver stage of a Plasmodium infection the parasite replicates from a single sporozoite into thousands of merozoites. Infection of humans and rodents with large numbers of sporozoites that arrest their development within the liver can cause sterile protection from subsequent infections. Disruption of genes essential for liver stage development of rodent malaria parasites has yielded a number of attenuated parasite strains. A key question to this end is how increased attenuation relates to vaccine efficacy. Here, we generated rodent malaria parasite lines that arrest during liver stage development and probed the impact of multiple gene deletions on attenuation and protective efficacy. In contrast to P. berghei strain ANKA LISP2(-) or uis3(-) single knockout parasites, which occasionally caused breakthrough infections, the double mutant lacking both genes was completely attenuated even when high numbers of sporozoites were administered. However, different vaccination protocols showed that LISP2(-) parasites protected better than uis3(-) and double mutants. Hence, deletion of several genes can yield increased safety but might come at the cost of protective efficacy.
Collapse
|
40
|
A Plasmodium yoelii Mei2-Like RNA Binding Protein Is Essential for Completion of Liver Stage Schizogony. Infect Immun 2016; 84:1336-1345. [PMID: 26883588 DOI: 10.1128/iai.01417-15] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 02/08/2016] [Indexed: 11/20/2022] Open
Abstract
Plasmodium parasites employ posttranscriptional regulatory mechanisms as their life cycle transitions between host cell invasion and replication within both the mosquito vector and mammalian host. RNA binding proteins (RBPs) provide one mechanism for modulation of RNA function. To explore the role of Plasmodium RBPs during parasite replication, we searched for RBPs that might play a role during liver stage development, the parasite stage that exhibits the most extensive growth and replication. We identified a parasite ortholog of the Mei2 (Meiosis inhibited 2) RBP that is conserved among Plasmodium species (PlasMei2) and exclusively transcribed in liver stage parasites. Epitope-tagged Plasmodium yoelii PlasMei2 was expressed only during liver stage schizogony and showed an apparent granular cytoplasmic location. Knockout of PlasMei2 (plasmei2(-)) in P. yoelii only affected late liver stage development. The P. yoelii plasmei2(-) liver stage size increased progressively until late in development, similar to wild-type parasite development. However, P. yoelii plasmei2(-) liver stage schizonts exhibited an abnormal DNA segregation phenotype and failed to form exoerythrocytic merozoites. Consequently the cellular integrity of P. yoelii plasmei2(-) liver stages became increasingly compromised late in development and the majority of P. yoelii plasmei2(-) underwent cell death by the time wild-type liver stages mature and release merozoites. This resulted in a complete block of P. yoelii plasmei2(-) transition from liver stage to blood stage infection in mice. Our results show for the first time the importance of a Plasmodium RBP in the coordinated progression of late liver stage schizogony and maturation of new invasive forms.
Collapse
|
41
|
Zhu L, Zhao J, Wang J, Hu C, Peng J, Luo R, Zhou C, Liu J, Lin J, Jin Y, Davis RE, Cheng G. MicroRNAs Are Involved in the Regulation of Ovary Development in the Pathogenic Blood Fluke Schistosoma japonicum. PLoS Pathog 2016; 12:e1005423. [PMID: 26871705 PMCID: PMC4752461 DOI: 10.1371/journal.ppat.1005423] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 01/07/2016] [Indexed: 12/19/2022] Open
Abstract
Schistosomes, blood flukes, are an important global public health concern. Paired adult female schistosomes produce large numbers of eggs that are primarily responsible for the disease pathology and critical for dissemination. Consequently, understanding schistosome sexual maturation and egg production may open novel perspectives for intervening with these processes to prevent clinical symptoms and to interrupt the life-cycle of these blood-flukes. microRNAs (miRNAs) are key regulators of many biological processes including development, cell proliferation, metabolism, and signal transduction. Here, we report on the identification of Schistosoma japonicum miRNAs using small RNA deep sequencing in the key stages of male-female pairing, gametogenesis, and egg production. We identified 38 miRNAs, including 10 previously unknown miRNAs. Eighteen of the miRNAs were differentially expressed between male and female schistosomes and during different stages of sexual maturation. We identified 30 potential target genes for 16 of the S. japonicum miRNAs using antibody-based pull-down assays and bioinformatic analyses. We further validated some of these target genes using either in vitro luciferase assays or in vivo miRNA suppression experiments. Notably, suppression of the female enriched miRNAs bantam and miR-31 led to morphological alteration of ovaries in female schistosomes. These findings uncover key roles for specific miRNAs in schistosome sexual maturation and egg production.
Collapse
Affiliation(s)
- Lihui Zhu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture, Beijing, China
| | - Jiangping Zhao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture, Beijing, China
| | - Jianbin Wang
- Departments of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Chao Hu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture, Beijing, China
| | - Jinbiao Peng
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture, Beijing, China
| | - Rong Luo
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture, Beijing, China
| | - Chunjing Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture, Beijing, China
| | - Juntao Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture, Beijing, China
| | - Jiaojiao Lin
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture, Beijing, China
| | - Youxin Jin
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Richard E. Davis
- Departments of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Guofeng Cheng
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Key Laboratory of Animal Parasitology, Ministry of Agriculture, Beijing, China
| |
Collapse
|
42
|
Silva PAGC, Guerreiro A, Santos JM, Braks JAM, Janse CJ, Mair GR. Translational Control of UIS4 Protein of the Host-Parasite Interface Is Mediated by the RNA Binding Protein Puf2 in Plasmodium berghei Sporozoites. PLoS One 2016; 11:e0147940. [PMID: 26808677 PMCID: PMC4726560 DOI: 10.1371/journal.pone.0147940] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 01/11/2016] [Indexed: 11/19/2022] Open
Abstract
UIS4 is a key protein component of the host-parasite interface in the liver stage of the rodent malaria parasite Plasmodium berghei and required for parasite survival after invasion. In the infectious sporozoite, UIS4 protein has variably been shown to be translated but also been reported to be translationally repressed. Here we show that uis4 mRNA translation is regulated by the P. berghei RNA binding protein Pumilio-2 (PbPuf2 or Puf2 from here on forward) in infectious salivary gland sporozoites in the mosquito vector. Using RNA immunoprecipitation we show that uis4 mRNA is bound by Puf2 in salivary gland sporozoites. In the absence of Puf2, uis4 mRNA translation is de-regulated and UIS4 protein expression upregulated in salivary gland sporozoites. Here, using RNA immunoprecipitation, we reveal the first Puf2-regulated mRNA in this parasite.
Collapse
Affiliation(s)
- Patrícia A. G. C. Silva
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, 1649–028, Lisbon, Portugal
| | - Ana Guerreiro
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, 1649–028, Lisbon, Portugal
| | - Jorge M. Santos
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, 1649–028, Lisbon, Portugal
| | | | | | - Gunnar R. Mair
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, 1649–028, Lisbon, Portugal
- Parasitology, Department of Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
43
|
Singer M, Marshall J, Heiss K, Mair GR, Grimm D, Mueller AK, Frischknecht F. Zinc finger nuclease-based double-strand breaks attenuate malaria parasites and reveal rare microhomology-mediated end joining. Genome Biol 2015; 16:249. [PMID: 26573820 PMCID: PMC4647826 DOI: 10.1186/s13059-015-0811-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/22/2015] [Indexed: 02/06/2023] Open
Abstract
Background Genome editing of malaria parasites is key to the generation of live attenuated parasites used in experimental vaccination approaches. DNA repair in Plasmodium generally occurs only through homologous recombination. This has been used to generate transgenic parasites that lack one to three genes, leading to developmental arrest in the liver and allowing the host to launch a protective immune response. While effective in principle, this approach is not safe for use in humans as single surviving parasites can still cause disease. Here we use zinc-finger nucleases to generate attenuated parasite lines lacking an entire chromosome arm, by a timed induction of a double-strand break. Rare surviving parasites also allow the investigation of unconventional DNA repair mechanisms in a rodent malaria parasite. Results A single, zinc-finger nuclease-induced DNA double-strand break results in the generation of attenuated parasite lines that show varying degrees of developmental arrest, protection efficacy in an immunisation regime and safety, depending on the timing of zinc-finger nuclease expression within the life cycle. We also identify DNA repair by microhomology-mediated end joining with as little as four base pairs, resulting in surviving parasites and thus breakthrough infections. Conclusions Malaria parasites can repair DNA double-strand breaks with surprisingly small mini-homology domains located across the break point. Timely expression of zinc-finger nucleases could be used to generate a new generation of attenuated parasite lines lacking hundreds of genes. Electronic supplementary material The online version of this article (doi:10.1186/s13059-015-0811-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mirko Singer
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany.
| | - Jennifer Marshall
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Kirsten Heiss
- Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany.,MalVa GmbH, Heidelberg, Germany
| | - Gunnar R Mair
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Dirk Grimm
- Virology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 267, 69120, Heidelberg, Germany
| | - Ann-Kristin Mueller
- Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany.,German Center for Infectious Diseases, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany.
| |
Collapse
|
44
|
Rijpma SR, van der Velden M, González-Pons M, Annoura T, van Schaijk BCL, van Gemert GJ, van den Heuvel JJMW, Ramesar J, Chevalley-Maurel S, Ploemen IH, Khan SM, Franetich JF, Mazier D, de Wilt JHW, Serrano AE, Russel FGM, Janse CJ, Sauerwein RW, Koenderink JB, Franke-Fayard BM. Multidrug ATP-binding cassette transporters are essential for hepatic development of Plasmodium sporozoites. Cell Microbiol 2015; 18:369-83. [PMID: 26332724 DOI: 10.1111/cmi.12517] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 08/11/2015] [Accepted: 08/24/2015] [Indexed: 12/23/2022]
Abstract
Multidrug resistance-associated proteins (MRPs) belong to the C-family of ATP-binding cassette (ABC) transport proteins and are known to transport a variety of physiologically important compounds and to be involved in the extrusion of pharmaceuticals. Rodent malaria parasites encode a single ABC transporter subfamily C protein, whereas human parasites encode two: MRP1 and MRP2. Although associated with drug resistance, their biological function and substrates remain unknown. To elucidate the role of MRP throughout the parasite life cycle, Plasmodium berghei and Plasmodium falciparum mutants lacking MRP expression were generated. P. berghei mutants lacking expression of the single MRP as well as P. falciparum mutants lacking MRP1, MRP2 or both proteins have similar blood stage growth kinetics and drug-sensitivity profiles as wild type parasites. We show that MRP1-deficient parasites readily invade primary human hepatocytes and develop into mature liver stages. In contrast, both P. falciparum MRP2-deficient parasites and P. berghei mutants lacking MRP protein expression abort in mid to late liver stage development, failing to produce mature liver stages. The combined P. berghei and P. falciparum data are the first demonstration of a critical role of an ABC transporter during Plasmodium liver stage development.
Collapse
Affiliation(s)
- Sanna R Rijpma
- Department of Pharmacology and Toxicology, Radboud University Medical Centre, Geert-Grooteplein 28, 6525 GA, Nijmegen, The Netherlands
| | - Maarten van der Velden
- Department of Pharmacology and Toxicology, Radboud University Medical Centre, Geert-Grooteplein 28, 6525 GA, Nijmegen, The Netherlands
| | - Maria González-Pons
- Department of Microbiology and Medical Zoology, University of Puerto Rico, School of Medicine, PR 00936-5067, San Juan, Puerto Rico, USA
| | - Takeshi Annoura
- Department of Tropical Medicine, The Jikei University School of Medicine, Post code 105-8461, Nishi-shinbashi 3-25-8, Minato-ku, Tokyo, Japan
| | - Ben C L van Schaijk
- Department of Medical Microbiology, Radboud University Medical Centre, Geert-Grooteplein 28, 6525 GA, Nijmegen, The Netherlands
| | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Medical Centre, Geert-Grooteplein 28, 6525 GA, Nijmegen, The Netherlands
| | - Jeroen J M W van den Heuvel
- Department of Pharmacology and Toxicology, Radboud University Medical Centre, Geert-Grooteplein 28, 6525 GA, Nijmegen, The Netherlands
| | - Jai Ramesar
- Department of Parasitology, Center of Infectious Diseases, Leiden Malaria Research Group, Leiden, The Netherlands
| | - Severine Chevalley-Maurel
- Department of Parasitology, Center of Infectious Diseases, Leiden Malaria Research Group, Leiden, The Netherlands
| | - Ivo H Ploemen
- Department of Medical Microbiology, Radboud University Medical Centre, Geert-Grooteplein 28, 6525 GA, Nijmegen, The Netherlands
| | - Shahid M Khan
- Department of Tropical Medicine, The Jikei University School of Medicine, Post code 105-8461, Nishi-shinbashi 3-25-8, Minato-ku, Tokyo, Japan
| | - Jean-Francois Franetich
- AP-HP, Groupe hospitalier Pitié-Salpêtrière, Service Parasitologie-Mycologie, 47-83 Boulevard de l'Hôpital, 75651, Paris, France
| | - Dominique Mazier
- AP-HP, Groupe hospitalier Pitié-Salpêtrière, Service Parasitologie-Mycologie, 47-83 Boulevard de l'Hôpital, 75651, Paris, France.,CIMI-Paris (UPMC UMRS CR7 - Inserm U1135 - CNRS ERL 8255), Paris, France
| | - Johannes H W de Wilt
- Department of Surgery, Radboud University Medical Centre, Geert Grooteplein 10, 6525 GA, Nijmegen, The Netherlands
| | - Adelfa E Serrano
- Department of Microbiology and Medical Zoology, University of Puerto Rico, School of Medicine, PR 00936-5067, San Juan, Puerto Rico, USA
| | - Frans G M Russel
- Department of Pharmacology and Toxicology, Radboud University Medical Centre, Geert-Grooteplein 28, 6525 GA, Nijmegen, The Netherlands
| | - Chris J Janse
- Department of Parasitology, Center of Infectious Diseases, Leiden Malaria Research Group, Leiden, The Netherlands
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud University Medical Centre, Geert-Grooteplein 28, 6525 GA, Nijmegen, The Netherlands
| | - Jan B Koenderink
- Department of Pharmacology and Toxicology, Radboud University Medical Centre, Geert-Grooteplein 28, 6525 GA, Nijmegen, The Netherlands
| | - Blandine M Franke-Fayard
- Department of Parasitology, Center of Infectious Diseases, Leiden Malaria Research Group, Leiden, The Netherlands
| |
Collapse
|
45
|
Bijker EM, Borrmann S, Kappe SH, Mordmüller B, Sack BK, Khan SM. Novel approaches to whole sporozoite vaccination against malaria. Vaccine 2015; 33:7462-8. [PMID: 26469716 PMCID: PMC6858867 DOI: 10.1016/j.vaccine.2015.09.095] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Revised: 08/22/2015] [Accepted: 09/22/2015] [Indexed: 12/15/2022]
Abstract
The parasitic disease malaria threatens more than 3 billion people worldwide, resulting in more than 200 million clinical cases and almost 600,000 deaths annually. Vaccines remain crucial for prevention and ultimately eradication of infectious diseases and, for malaria, whole sporozoite based immunization has been shown to be the most effective in experimental settings. In addition to immunization with radiation-attenuated sporozoites, chemoprophylaxis and sporozoites (CPS) is a highly efficient strategy to induce sterile protection in humans. Genetically attenuated parasites (GAP) have demonstrated significant protection in rodent studies, and are now being advanced into clinical testing. This review describes the existing pre-clinical and clinical data on CPS and GAP, discusses recent developments and examines how to transform these immunization approaches into vaccine candidates for clinical development.
Collapse
Affiliation(s)
- Else M Bijker
- Radboud University Medical Center, Department of Medical Microbiology, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| | - Steffen Borrmann
- Institute for Tropical Medicine, University of Tübingen, Tübingen, Germany; German Centre for Infection Research, University of Tübingen, Tübingen, Germany; Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Stefan H Kappe
- Seattle Biomedical Research Institute, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA
| | - Benjamin Mordmüller
- Institute for Tropical Medicine, University of Tübingen, Tübingen, Germany; German Centre for Infection Research, University of Tübingen, Tübingen, Germany; Centre de Recherches Médicales de Lambaréné, Alberts Schweitzer Hospital, BP 118 Lambaréné, Gabon
| | | | - Shahid M Khan
- Leiden University Medical Center, Department of Parasitology, PO Box 9600, 2300 RC Leiden, The Netherlands
| |
Collapse
|
46
|
Host cell phosphatidylcholine is a key mediator of malaria parasite survival during liver stage infection. Cell Host Microbe 2015; 16:778-86. [PMID: 25498345 PMCID: PMC4271766 DOI: 10.1016/j.chom.2014.11.006] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 09/29/2014] [Accepted: 11/04/2014] [Indexed: 01/22/2023]
Abstract
During invasion, Plasmodium, the causative agent of malaria, wraps itself in a parasitophorous vacuole membrane (PVM), which constitutes a critical interface between the parasite and its host cell. Within hepatocytes, each Plasmodium sporozoite generates thousands of new parasites, creating high demand for lipids to support this replication and enlarge the PVM. Here, a global analysis of the total lipid repertoire of Plasmodium-infected hepatocytes reveals an enrichment of neutral lipids and the major membrane phospholipid, phosphatidylcholine (PC). While infection is unaffected in mice deficient in key enzymes involved in neutral lipid synthesis and lipolysis, ablation of rate-limiting enzymes in hepatic PC biosynthetic pathways significantly decreases parasite numbers. Host PC is taken up by both P. berghei and P. falciparum and is necessary for correct localization of parasite proteins to the PVM, which is essential for parasite survival. Thus, Plasmodium relies on the abundance of these lipids within hepatocytes to support infection.
Collapse
|
47
|
Kumar H, Frischknecht F, Mair GR, Gomes J. In silico identification of genetically attenuated vaccine candidate genes for Plasmodium liver stage. INFECTION GENETICS AND EVOLUTION 2015; 36:72-81. [PMID: 26348884 DOI: 10.1016/j.meegid.2015.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 08/28/2015] [Accepted: 09/02/2015] [Indexed: 12/28/2022]
Abstract
Genetically attenuated parasites (GAPs) that lack genes essential for the liver stage of the malaria parasite, and therefore cause developmental arrest, have been developed as live vaccines in rodent malaria models and recently been tested in humans. The genes targeted for deletion were often identified by trial and error. Here we present a systematic gene - protein and transcript - expression analyses of several Plasmodium species with the aim to identify candidate genes for the generation of novel GAPs. With a lack of liver stage expression data for human malaria parasites, we used data available for liver stage development of Plasmodium yoelii, a rodent malaria model, to identify proteins expressed in the liver stage but absent from blood stage parasites. An orthology-based search was then employed to identify orthologous proteins in the human malaria parasite Plasmodium falciparum resulting in a total of 310 genes expressed in the liver stage but lacking evidence of protein expression in blood stage parasites. Among these 310 possible GAP candidates, we further studied Plasmodium liver stage proteins by phyletic distribution and functional domain analyses and shortlisted twenty GAP-candidates; these are: fabB/F, fabI, arp, 3 genes encoding subunits of the PDH complex, dnaJ, urm1, rS5, ancp, mcp, arh, gk, lisp2, valS, palm, and four conserved Plasmodium proteins of unknown function. Parasites lacking one or several of these genes might yield new attenuated malaria parasites for experimental vaccination studies.
Collapse
Affiliation(s)
- Hirdesh Kumar
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi 110016, India.; Integrative Parasitology, Department of Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Friedrich Frischknecht
- Integrative Parasitology, Department of Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Gunnar R Mair
- Integrative Parasitology, Department of Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - James Gomes
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi 110016, India..
| |
Collapse
|
48
|
The Plasmodium berghei translocon of exported proteins reveals spatiotemporal dynamics of tubular extensions. Sci Rep 2015. [PMID: 26219962 PMCID: PMC4518229 DOI: 10.1038/srep12532] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The erythrocyte is an extraordinary host cell for intracellular pathogens and requires extensive remodelling to become permissive for infection. Malaria parasites modify their host red blood cells through protein export to acquire nutrients and evade immune responses. Endogenous fluorescent tagging of three signature proteins of the Plasmodium berghei translocon of exported proteins (PTEX), heat shock protein 101, exported protein 2 (EXP2), and PTEX88, revealed motile, tubular extensions of the parasitophorous vacuole that protrude from the parasite far into the red blood cell. EXP2 displays a more prominent presence at the periphery of the parasite, consistent with its proposed role in pore formation. The tubular compartment is most prominent during trophozoite growth. Distinct spatiotemporal expression of individual PTEX components during sporogony and liver-stage development indicates additional functions and tight regulation of the PTEX translocon during parasite life cycle progression. Together, live cell imaging and correlative light and electron microscopy permitted previously unrecognized spatiotemporal and subcellular resolution of PTEX-containing tubules in murine malaria parasites. These findings further refine current models for Plasmodium-induced erythrocyte makeover.
Collapse
|
49
|
Jaijyan DK, Singh H, Singh AP. A Sporozoite- and Liver Stage-expressed Tryptophan-rich Protein Plays an Auxiliary Role in Plasmodium Liver Stage Development and Is a Potential Vaccine Candidate. J Biol Chem 2015; 290:19496-511. [PMID: 25960542 DOI: 10.1074/jbc.m114.588129] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Indexed: 01/07/2023] Open
Abstract
The liver stages of the malaria parasite are clinically silent and constitute ideal targets for causal prophylactic drugs and vaccines. Cellular and molecular events responsible for liver stage development are poorly characterized. Here, we show that sporozoite, liver stage tryptophan-rich protein (SLTRiP) forms large multimers. Mice immunized with a purified recombinant SLTRiP protein gave high antibody titers in both inbred and outbred mice. Immunized mice showed highly significant levels of protection upon challenge with sporozoites and exhibited 10,000-fold fewer parasite 18S-rRNA copy numbers in their livers. The protection offered by immunization with SLTRiP came mainly from T-cells, and antibodies had little role to play despite their high titers. Immunofluorescence assays showed that SLTRiP is expressed in the sporozoite and early to late liver stages of malaria parasites. SLTRiP protein is exported to the cytosol of infected host cells during the early hours of parasite infection. Parasites deficient in SLTRiP were moderately defective in liver stage parasite development. A transcriptome profile of SLTRiP-deficient parasite-infected hepatocytes highlighted that SLTRiP interferes with multiple pathways in the host cell. We have demonstrated a role for SLTRiP in sporozoites and the liver stage of malaria parasites.
Collapse
Affiliation(s)
- Dabbu Kumar Jaijyan
- From the Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Himanshu Singh
- From the Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Agam Prasad Singh
- From the Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| |
Collapse
|
50
|
In Vivo Function of PTEX88 in Malaria Parasite Sequestration and Virulence. EUKARYOTIC CELL 2015; 14:528-34. [PMID: 25820521 DOI: 10.1128/ec.00276-14] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 03/25/2015] [Indexed: 01/07/2023]
Abstract
Malaria pathology is linked to remodeling of red blood cells by eukaryotic Plasmodium parasites. Central to host cell refurbishment is the trafficking of parasite-encoded virulence factors through the Plasmodium translocon of exported proteins (PTEX). Much of our understanding of its function is based on experimental work with cultured Plasmodium falciparum, yet direct consequences of PTEX impairment during an infection remain poorly defined. Using the murine malaria model parasite Plasmodium berghei, it is shown here that efficient sequestration to the pulmonary, adipose, and brain tissue vasculature is dependent on the PTEX components thioredoxin 2 (TRX2) and PTEX88. While TRX2-deficient parasites remain virulent, PTEX88-deficient parasites no longer sequester in the brain, correlating with abolishment of cerebral complications in infected mice. However, an apparent trade-off for virulence attenuation was spleen enlargement, which correlates with a strongly reduced schizont-to-ring-stage transition. Strikingly, general protein export is unaffected in PTEX88-deficient mutants that mature normally in vitro. Thus, PTEX88 is pivotal for tissue sequestration in vivo, parasite virulence, and preventing exacerbation of spleen pathology, but these functions do not correlate with general protein export to the host erythrocyte. The presented data suggest that the protein export machinery of Plasmodium parasites and their underlying mechanistic features are considerably more complex than previously anticipated and indicate challenges for targeted intervention strategies.
Collapse
|