51
|
Andersson AMC, Buldun CM, Pattinson DJ, Draper SJ, Howarth M. SnoopLigase peptide-peptide conjugation enables modular vaccine assembly. Sci Rep 2019; 9:4625. [PMID: 30874593 PMCID: PMC6420506 DOI: 10.1038/s41598-019-40985-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 02/11/2019] [Indexed: 02/06/2023] Open
Abstract
For many infectious diseases there is still no vaccine, even though potential protective antigens have been identified. Suitable platforms and conjugation routes are urgently needed to convert the promise of such antigens into broadly protective and scalable vaccines. Here we apply a newly established peptide-peptide ligation approach, SnoopLigase, for specific and irreversible coupling of antigens onto an oligomerization platform. SnoopLigase was engineered from a Streptococcus pneumoniae adhesin and enables isopeptide bond formation between two peptide tags: DogTag and SnoopTagJr. We expressed in bacteria DogTag linked to the self-assembling coiled-coil nanoparticle IMX313. This platform was stable over months at 37 °C when lyophilized, remaining reactive even after boiling. IMX-DogTag was efficiently coupled to two blood-stage malarial proteins (from PfEMP1 or CyRPA), with SnoopTagJr fused at the N- or C-terminus. We also showed SnoopLigase-mediated coupling of a telomerase peptide relevant to cancer immunotherapy. SnoopLigase-mediated nanoassembly enhanced the antibody response to both malaria antigens in a prime-boost model. Including or depleting SnoopLigase from the conjugate had little effect on the antibody response to the malarial antigens. SnoopLigase decoration represents a promising and accessible strategy for modular plug-and-display vaccine assembly, as well as providing opportunities for robust nanoconstruction in synthetic biology.
Collapse
Affiliation(s)
| | - Can M Buldun
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | | | - Simon J Draper
- Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Mark Howarth
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK.
| |
Collapse
|
52
|
Abstract
The development of highly effective and durable vaccines against the human malaria parasites Plasmodium falciparum and P. vivax remains a key priority. Decades of endeavor have taught that achieving this goal will be challenging; however, recent innovation in malaria vaccine research and a diverse pipeline of novel vaccine candidates for clinical assessment provides optimism. With first-generation pre-erythrocytic vaccines aiming for licensure in the coming years, it is important to reflect on how next-generation approaches can improve on their success. Here we review the latest vaccine approaches that seek to prevent malaria infection, disease, and transmission and highlight some of the major underlying immunological and molecular mechanisms of protection. The synthesis of rational antigen selection, immunogen design, and immunization strategies to induce quantitatively and qualitatively improved immune effector mechanisms offers promise for achieving sustained high-level protection.
Collapse
|
53
|
Tan J, Piccoli L, Lanzavecchia A. The Antibody Response to Plasmodium falciparum: Cues for Vaccine Design and the Discovery of Receptor-Based Antibodies. Annu Rev Immunol 2018; 37:225-246. [PMID: 30566366 DOI: 10.1146/annurev-immunol-042617-053301] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Plasmodium falciparum remains a serious public health problem and a continuous challenge for the immune system due to the complexity and diversity of the pathogen. Recent advances from several laboratories in the characterization of the antibody response to the parasite have led to the identification of critical targets for protection and revealed a new mechanism of diversification based on the insertion of host receptors into immunoglobulin genes, leading to the production of receptor-based antibodies. These advances have opened new possibilities for vaccine design and passive antibody therapies to provide sterilizing immunity and control blood-stage parasites.
Collapse
Affiliation(s)
- Joshua Tan
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland; .,Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, United Kingdom.,Current affiliation: National Institute of Allergy and Infectious Diseases, Rockville, Maryland 20852, USA
| | - Luca Piccoli
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland;
| | - Antonio Lanzavecchia
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland; .,VIR Biotechnology, San Francisco, California 94158, USA
| |
Collapse
|
54
|
Wong W, Huang R, Menant S, Hong C, Sandow JJ, Birkinshaw RW, Healer J, Hodder AN, Kanjee U, Tonkin CJ, Heckmann D, Soroka V, Søgaard TMM, Jørgensen T, Duraisingh MT, Czabotar PE, de Jongh WA, Tham WH, Webb AI, Yu Z, Cowman AF. Structure of Plasmodium falciparum Rh5-CyRPA-Ripr invasion complex. Nature 2018; 565:118-121. [PMID: 30542156 DOI: 10.1038/s41586-018-0779-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 10/25/2018] [Indexed: 01/24/2023]
Abstract
Plasmodium falciparum causes the severe form of malaria that has high levels of mortality in humans. Blood-stage merozoites of P. falciparum invade erythrocytes, and this requires interactions between multiple ligands from the parasite and receptors in hosts. These interactions include the binding of the Rh5-CyRPA-Ripr complex with the erythrocyte receptor basigin1,2, which is an essential step for entry into human erythrocytes. Here we show that the Rh5-CyRPA-Ripr complex binds the erythrocyte cell line JK-1 significantly better than does Rh5 alone, and that this binding occurs through the insertion of Rh5 and Ripr into host membranes as a complex with high molecular weight. We report a cryo-electron microscopy structure of the Rh5-CyRPA-Ripr complex at subnanometre resolution, which reveals the organization of this essential invasion complex and the mode of interactions between members of the complex, and shows that CyRPA is a critical mediator of complex assembly. Our structure identifies blades 4-6 of the β-propeller of CyRPA as contact sites for Rh5 and Ripr. The limited contacts between Rh5-CyRPA and CyRPA-Ripr are consistent with the dissociation of Rh5 and Ripr from CyRPA for membrane insertion. A comparision of the crystal structure of Rh5-basigin with the cryo-electron microscopy structure of Rh5-CyRPA-Ripr suggests that Rh5 and Ripr are positioned parallel to the erythrocyte membrane before membrane insertion. This provides information on the function of this complex, and thereby provides insights into invasion by P. falciparum.
Collapse
Affiliation(s)
- Wilson Wong
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Rick Huang
- CryoEM Shared Resources, Janelia Farm Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Sebastien Menant
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Chuan Hong
- CryoEM Shared Resources, Janelia Farm Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Jarrod J Sandow
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Richard W Birkinshaw
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Julie Healer
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Anthony N Hodder
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Usheer Kanjee
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Christopher J Tonkin
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Denise Heckmann
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | | | | | | | - Manoj T Duraisingh
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Peter E Czabotar
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | | | - Wai-Hong Tham
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Andrew I Webb
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Zhiheng Yu
- CryoEM Shared Resources, Janelia Farm Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Alan F Cowman
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. .,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
55
|
Quintana MDP, Ch’ng JH, Zandian A, Imam M, Hultenby K, Theisen M, Nilsson P, Qundos U, Moll K, Chan S, Wahlgren M. SURGE complex of Plasmodium falciparum in the rhoptry-neck (SURFIN4.2-RON4-GLURP) contributes to merozoite invasion. PLoS One 2018; 13:e0201669. [PMID: 30092030 PMCID: PMC6084945 DOI: 10.1371/journal.pone.0201669] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 07/19/2018] [Indexed: 12/25/2022] Open
Abstract
Plasmodium falciparum invasion into red blood cells (RBCs) is a complex process engaging proteins on the merozoite surface and those contained and sequentially released from the apical organelles (micronemes and rhoptries). Fundamental to invasion is the formation of a moving junction (MJ), a region of close apposition of the merozoite and the RBC plasma membranes, through which the merozoite draws itself before settling into a newly formed parasitophorous vacuole (PV). SURFIN4.2 was identified at the surface of the parasitized RBCs (pRBCs) but was also found apically associated with the merozoite. Using antibodies against the N-terminus of the protein we show the presence of SURFIN4.2 in the neck of the rhoptries, its secretion into the PV and shedding into the culture supernatant upon schizont rupture. Using immunoprecipitation followed by mass spectrometry we describe here a novel protein complex we have named SURGE where SURFIN4.2 forms interacts with the rhoptry neck protein 4 (RON4) and the Glutamate Rich Protein (GLURP). The N-terminal cysteine-rich-domain (CRD) of SURFIN4.2 mediates binding to the RBC membrane and its interaction with RON4 suggests its involvement in the contact between the merozoite apex and the RBC at the MJ. Supporting this suggestion, we also found that polyclonal antibodies to the extracellular domain (including the CRD) of SURFIN4.2 partially inhibit merozoite invasion. We propose that the formation of the SURGE complex participates in the establishment of parasite infection within the PV and the RBCs.
Collapse
Affiliation(s)
- Maria del Pilar Quintana
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, Stockholm, Sweden
| | - Jun-Hong Ch’ng
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, Stockholm, Sweden
- Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Arash Zandian
- Affinity Proteomics, Science for Life Laboratory, School of Biotechnology, KTH-Royal Institutet of Technology, Stockholm, Sweden
| | - Maryam Imam
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, Stockholm, Sweden
| | - Kjell Hultenby
- Division of Clinical Research Centre, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Centre for Medical Parasitology, Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Peter Nilsson
- Affinity Proteomics, Science for Life Laboratory, School of Biotechnology, KTH-Royal Institutet of Technology, Stockholm, Sweden
| | - Ulrika Qundos
- Affinity Proteomics, Science for Life Laboratory, School of Biotechnology, KTH-Royal Institutet of Technology, Stockholm, Sweden
| | - Kirsten Moll
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, Stockholm, Sweden
| | - Sherwin Chan
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, Stockholm, Sweden
| | - Mats Wahlgren
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
56
|
Kinetics of antibody responses to PfRH5-complex antigens in Ghanaian children with Plasmodium falciparum malaria. PLoS One 2018; 13:e0198371. [PMID: 29883485 PMCID: PMC5993283 DOI: 10.1371/journal.pone.0198371] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/17/2018] [Indexed: 11/21/2022] Open
Abstract
Plasmodium falciparum PfRH5 protein binds Ripr, CyRPA and Pf113 to form a complex that is essential for merozoite invasion of erythrocytes. The inter-genomic conservation of the PfRH5 complex proteins makes them attractive blood stage vaccine candidates. However, little is known about how antibodies to PfRH5, CyRPA and Pf113 are acquired and maintained in naturally exposed populations, and the role of PfRH5 complex proteins in naturally acquired immunity. To provide such data, we studied 206 Ghanaian children between the ages of 1–12 years, who were symptomatic, asymptomatic or aparasitemic and healthy. Plasma levels of antigen-specific IgG and IgG subclasses were measured by ELISA at several time points during acute disease and convalescence. On the day of admission with acute P. falciparum malaria, the prevalence of antibodies to PfRH5-complex proteins was low compared to other merozoite antigens (EBA175, GLURP-R0 and GLURP-R2). At convalescence, the levels of RH5-complex-specific IgG were reduced, with the decay of PfRH5-specific IgG being slower than the decay of IgG specific for CyRPA and Pf113. No correlation between IgG levels and protection against P. falciparum malaria was observed for any of the PfRH5 complex proteins. From this we conclude that specific IgG was induced against proteins from the PfRH5-complex during acute P. falciparum malaria, but the prevalence was low and the IgG levels decayed rapidly after treatment. These data indicate that the levels of IgG specific for PfRH5-complex proteins in natural infections in Ghanaian children were markers of recent exposure only.
Collapse
|
57
|
Pino P, Caldelari R, Mukherjee B, Vahokoski J, Klages N, Maco B, Collins CR, Blackman MJ, Kursula I, Heussler V, Brochet M, Soldati-Favre D. A multistage antimalarial targets the plasmepsins IX and X essential for invasion and egress. Science 2018; 358:522-528. [PMID: 29074775 DOI: 10.1126/science.aaf8675] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 09/18/2017] [Indexed: 12/20/2022]
Abstract
Regulated exocytosis by secretory organelles is important for malaria parasite invasion and egress. Many parasite effector proteins, including perforins, adhesins, and proteases, are extensively proteolytically processed both pre- and postexocytosis. Here we report the multistage antiplasmodial activity of the aspartic protease inhibitor hydroxyl-ethyl-amine-based scaffold compound 49c. This scaffold inhibits the preexocytosis processing of several secreted rhoptry and microneme proteins by targeting the corresponding maturases plasmepsins IX (PMIX) and X (PMX), respectively. Conditional excision of PMIX revealed its crucial role in invasion, and recombinantly active PMIX and PMX cleave egress and invasion factors in a 49c-sensitive manner.
Collapse
Affiliation(s)
- Paco Pino
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva, Centre Médical Universitaire (CMU), 1211 Geneva, Switzerland.
| | - Reto Caldelari
- Institute of Cell Biology, University of Bern, 3012 Bern, Switzerland
| | - Budhaditya Mukherjee
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva, Centre Médical Universitaire (CMU), 1211 Geneva, Switzerland
| | - Juha Vahokoski
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Natacha Klages
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva, Centre Médical Universitaire (CMU), 1211 Geneva, Switzerland
| | - Bohumil Maco
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva, Centre Médical Universitaire (CMU), 1211 Geneva, Switzerland
| | - Christine R Collins
- Malaria Biochemistry Laboratory, The Francis Crick Institute, Mill Hill, London NW1 1AT, UK
| | - Michael J Blackman
- Malaria Biochemistry Laboratory, The Francis Crick Institute, Mill Hill, London NW1 1AT, UK.,Department of Pathogen Molecular Biology, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Inari Kursula
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway.,Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 7, 90220 Oulu, Finland
| | - Volker Heussler
- Institute of Cell Biology, University of Bern, 3012 Bern, Switzerland
| | - Mathieu Brochet
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva, Centre Médical Universitaire (CMU), 1211 Geneva, Switzerland
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva, Centre Médical Universitaire (CMU), 1211 Geneva, Switzerland.
| |
Collapse
|
58
|
Abstract
Plasmodium species cause malaria by proliferating in human erythrocytes. Invasion of immunologically privileged erythrocytes provides a relatively protective niche as well as access to a rich source of nutrients. Plasmodium spp. target erythrocytes of different ages, but share a common mechanism of invasion. Specific engagement of erythrocyte receptors defines target cell tropism, activating downstream events and resulting in the physical penetration of the erythrocyte, powered by the parasite's actinomyosin-based motor. Here we review the latest in our understanding of the molecular composition of this highly complex and fascinating biological process.
Collapse
|
59
|
Plenderleith LJ, Liu W, MacLean OA, Li Y, Loy DE, Sundararaman SA, Bibollet-Ruche F, Learn GH, Hahn BH, Sharp PM. Adaptive Evolution of RH5 in Ape Plasmodium species of the Laverania Subgenus. mBio 2018; 9:e02237-17. [PMID: 29362238 PMCID: PMC5784257 DOI: 10.1128/mbio.02237-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 12/11/2017] [Indexed: 01/03/2023] Open
Abstract
Plasmodium falciparum, the major cause of malaria morbidity and mortality in humans, has been shown to have emerged after cross-species transmission of one of six host-specific parasites (subgenus Laverania) infecting wild chimpanzees (Pan troglodytes) and western gorillas (Gorilla gorilla). Binding of the parasite-encoded ligand RH5 to the host protein basigin is essential for erythrocyte invasion and has been implicated in host specificity. A recent study claimed to have found two amino acid changes in RH5 that "drove the host shift leading to the emergence of P. falciparum as a human pathogen." However, the ape Laverania data available at that time, which included only a single distantly related chimpanzee parasite sequence, were inadequate to justify any such conclusion. Here, we have investigated Laverania Rh5 gene evolution using sequences from all six ape parasite species. Searching for gene-wide episodic selection across the entire Laverania phylogeny, we found eight codons to be under positive selection, including three that correspond to contact residues known to form hydrogen bonds between P. falciparum RH5 and human basigin. One of these sites (residue 197) has changed subsequent to the transmission from apes to humans that gave rise to P. falciparum, suggesting a possible role in the adaptation of the gorilla parasite to the human host. We also found evidence that the patterns of nucleotide polymorphisms in P. falciparum are not typical of Laverania species and likely reflect the recent demographic history of the human parasite.IMPORTANCE A number of closely related, host-specific malaria parasites infecting wild chimpanzees and gorillas have recently been described. The most important cause of human malaria, Plasmodium falciparum, is now known to have resulted from a cross-species transmission of one of the gorilla parasites. Overcoming species-specific interactions between a parasite ligand, RH5, and its receptor on host cells, basigin, was likely an important step in the origin of the human parasite. We have investigated the evolution of the Rh5 gene and found evidence of adaptive changes during the diversification of the ape parasite species at sites that are known to form bonds with human basigin. One of these changes occurred at the origin of P. falciparum, implicating it as an important adaptation to the human host.
Collapse
Affiliation(s)
- Lindsey J Plenderleith
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Immunity, Infection and Evolution, University of Edinburgh, Edinburgh, United Kingdom
| | - Weimin Liu
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Oscar A MacLean
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Immunity, Infection and Evolution, University of Edinburgh, Edinburgh, United Kingdom
| | - Yingying Li
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dorothy E Loy
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sesh A Sundararaman
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Gerald H Learn
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Beatrice H Hahn
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paul M Sharp
- Institute of Evolutionary Biology, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Immunity, Infection and Evolution, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
60
|
Mechanisms of naturally acquired immunity to P. falciparum and approaches to identify merozoite antigen targets. Parasitology 2017; 145:839-847. [PMID: 29144217 DOI: 10.1017/s0031182017001949] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Malaria is one the most serious infectious diseases with over 200 million clinical cases annually. Most cases of the severe disease are caused by Plasmodium falciparum. The blood stage of Plasmodium parasite is entirely responsible for malaria-associated pathology. The population most susceptible to severe malaria are children under the age of 5, with low levels of immunity. It is only after many years of repeated exposure that individuals living in endemic areas develop clinical immunity. This form of protection prevents clinical episodes by substantially reducing parasite burden. Naturally acquired immunity predominantly targets blood-stage parasites with antibody responses being the main mediators of protection. The targets of clinical immunity are the extracellular merozoite and the infected erythrocyte surface, with the extremely diverse PfEMP1 proteins the main target here. This observation provides a strong rationale that an effective anti-malaria vaccine targeting blood-stage parasites is achievable. Thus the identification of antigenic targets of naturally acquired immunity remains an important step towards the formulation of novel vaccine combinations before testing their efficacy in clinical trials. This review summarizes the main findings to date defining antigenic targets present on the extracellular merozoite associated with naturally acquired immunity to P. falciparum malaria.
Collapse
|
61
|
Payne RO, Silk SE, Elias SC, Miura K, Diouf A, Galaway F, de Graaf H, Brendish NJ, Poulton ID, Griffiths OJ, Edwards NJ, Jin J, Labbé GM, Alanine DG, Siani L, Di Marco S, Roberts R, Green N, Berrie E, Ishizuka AS, Nielsen CM, Bardelli M, Partey FD, Ofori MF, Barfod L, Wambua J, Murungi LM, Osier FH, Biswas S, McCarthy JS, Minassian AM, Ashfield R, Viebig NK, Nugent FL, Douglas AD, Vekemans J, Wright GJ, Faust SN, Hill AV, Long CA, Lawrie AM, Draper SJ. Human vaccination against RH5 induces neutralizing antimalarial antibodies that inhibit RH5 invasion complex interactions. JCI Insight 2017; 2:96381. [PMID: 29093263 PMCID: PMC5752323 DOI: 10.1172/jci.insight.96381] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/05/2017] [Indexed: 11/17/2022] Open
Abstract
The development of a highly effective vaccine remains a key strategic goal to aid the control and eventual eradication of Plasmodium falciparum malaria. In recent years, the reticulocyte-binding protein homolog 5 (RH5) has emerged as the most promising blood-stage P. falciparum candidate antigen to date, capable of conferring protection against stringent challenge in Aotus monkeys. We report on the first clinical trial to our knowledge to assess the RH5 antigen - a dose-escalation phase Ia study in 24 healthy, malaria-naive adult volunteers. We utilized established viral vectors, the replication-deficient chimpanzee adenovirus serotype 63 (ChAd63), and the attenuated orthopoxvirus modified vaccinia virus Ankara (MVA), encoding RH5 from the 3D7 clone of P. falciparum. Vaccines were administered i.m. in a heterologous prime-boost regimen using an 8-week interval and were well tolerated. Vaccine-induced anti-RH5 serum antibodies exhibited cross-strain functional growth inhibition activity (GIA) in vitro, targeted linear and conformational epitopes within RH5, and inhibited key interactions within the RH5 invasion complex. This is the first time to our knowledge that substantial RH5-specific responses have been induced by immunization in humans, with levels greatly exceeding the serum antibody responses observed in African adults following years of natural malaria exposure. These data support the progression of RH5-based vaccines to human efficacy testing.
Collapse
Affiliation(s)
- Ruth O. Payne
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Sarah E. Silk
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Sean C. Elias
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, Maryland, USA
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, Maryland, USA
| | - Francis Galaway
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Hans de Graaf
- NIHR Wellcome Trust Clinical Research Facility, University Hospital Southampton NHS Foundation Trust and Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Nathan J. Brendish
- NIHR Wellcome Trust Clinical Research Facility, University Hospital Southampton NHS Foundation Trust and Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Ian D. Poulton
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Nick J. Edwards
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Jing Jin
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | | - Loredana Siani
- ReiThera SRL (formerly Okairos SRL), Viale Città d’Europa, Rome, Italy
| | - Stefania Di Marco
- ReiThera SRL (formerly Okairos SRL), Viale Città d’Europa, Rome, Italy
| | - Rachel Roberts
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Nicky Green
- Clinical Biomanufacturing Facility, University of Oxford, Oxford, United Kingdom
| | - Eleanor Berrie
- Clinical Biomanufacturing Facility, University of Oxford, Oxford, United Kingdom
| | | | | | - Martino Bardelli
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Frederica D. Partey
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
- Centre for Medical Parasitology, Department of Immunology and Microbiology (ISIM), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Michael F. Ofori
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Lea Barfod
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Juliana Wambua
- KEMRI Centre for Geographic Medicine Research, Kilifi, Kenya
| | - Linda M. Murungi
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
- KEMRI Centre for Geographic Medicine Research, Kilifi, Kenya
| | - Faith H. Osier
- KEMRI Centre for Geographic Medicine Research, Kilifi, Kenya
| | - Sumi Biswas
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - James S. McCarthy
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | | | - Rebecca Ashfield
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Nicola K. Viebig
- European Vaccine Initiative, UniversitätsKlinikum Heidelberg, Heidelberg, Germany
| | - Fay L. Nugent
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | | - Gavin J. Wright
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Saul N. Faust
- NIHR Wellcome Trust Clinical Research Facility, University Hospital Southampton NHS Foundation Trust and Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Adrian V.S. Hill
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Carole A. Long
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, Maryland, USA
| | - Alison M. Lawrie
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Simon J. Draper
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
62
|
Mother-Newborn Pairs in Malawi Have Similar Antibody Repertoires to Diverse Malaria Antigens. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00136-17. [PMID: 28835359 DOI: 10.1128/cvi.00136-17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 07/31/2017] [Indexed: 11/20/2022]
Abstract
Maternal antibodies may play a role in protecting newborns against malaria disease. Plasmodium falciparum parasite surface antigens are diverse, and protection from infection requires allele-specific immunity. Although malaria-specific antibodies have been shown to cross the placenta, the extent to which antibodies that respond to the full repertoire of diverse antigens are transferred from the mother to the infant has not been explored. Understanding the breadth of maternal antibody responses and to what extent these antibodies are transferred to the child can inform vaccine design and evaluation. We probed plasma from cord blood and serum from mothers at delivery using a customized protein microarray that included variants of malaria vaccine target antigens to assess the intensity and breadth of seroreactivity to three malaria vaccine candidate antigens in mother-newborn pairs in Malawi. Among the 33 paired specimens that were assessed, mothers and newborns had similar intensity and repertoire of seroreactivity. Maternal antibody levels against vaccine candidate antigens were the strongest predictors of infant antibody levels. Placental malaria did not significantly impair transplacental antibody transfer. However, mothers with placental malaria had significantly higher antibody levels against these blood-stage antigens than mothers without placental malaria. The repertoire and levels of infant antibodies against a wide range of malaria vaccine candidate antigen variants closely mirror maternal levels in breadth and magnitude regardless of evidence of placental malaria. Vaccinating mothers with an effective malaria vaccine during pregnancy may induce high and potentially protective antibody repertoires in newborns.
Collapse
|
63
|
Fotoran WL, Santangelo R, de Miranda BNM, Irvine DJ, Wunderlich G. DNA-Loaded Cationic Liposomes Efficiently Function as a Vaccine against Malarial Proteins. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 7:1-10. [PMID: 28879213 PMCID: PMC5581859 DOI: 10.1016/j.omtm.2017.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 08/18/2017] [Indexed: 12/14/2022]
Abstract
The delivery of antigens as DNA vaccines is an efficient alternative to induce immune responses against antigens, which are difficult to produce in recombinant form. However, the delivery of naked DNA is ineffective or relies on sophisticated ballistic devices. Here, we show a combination of liposome application and naked DNA vaccine that successfully overcomes these problems. Upon entrapment of plasmids encoding different antigens in cationic particles, transfection efficiencies similar to commercial kits were achieved in in vitro cell cultures. The liposome-based approach provided strong humoral responses against three malarial antigens, namely the Circumsporozoite protein and the C terminus of merozoite surface protein 1 from Plasmodium vivax (titers 104 or 103–104, respectively) and P. falciparum Rhoptry antigen 5 from Plasmodium falciparum (titers 103–104). When employed in P. falciparum growth-inhibition assays, antibodies demonstrated consistent reinvasion-blocking activities that were dose dependent. Liposome-formulated DNA vaccines may prove useful when targets cannot be produced as recombinant proteins and when conformation-dependent and highly specific antibodies are mandatory.
Collapse
Affiliation(s)
- Wesley L Fotoran
- Department of Parasitology, Institute for Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes 1374, São Paulo, 05508000, Brazil
| | - Rachele Santangelo
- Department of Parasitology, Institute for Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes 1374, São Paulo, 05508000, Brazil
| | - Beatriz N M de Miranda
- Institute of Chemistry of São Carlos, University of São Paulo, Av. Trabalhador São-Carlense 400, São Carlos, 13566-590, Brazil
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research and Department of Biomedical Engineering and Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Ragon Institute of MGH, MIT and Harvard University, Boston, MA, USA.,Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Gerhard Wunderlich
- Department of Parasitology, Institute for Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes 1374, São Paulo, 05508000, Brazil
| |
Collapse
|
64
|
Abstract
Evidence accumulated through the years clearly indicates that antiparasite immune responses can efficiently control malaria parasite infection at all development stages, and under certain circumstances they can prevent parasite infection. Translating these findings into vaccines or immunotherapeutic interventions has been difficult in part because of the extraordinary biological complexity of this parasite, which has several developmental stages expressing unique sets of stage-specific genes and multiple antigens, most of which are antigenically diverse. Nevertheless, in the last 30 years major advances have resulted in characterization of a number of vaccine candidates, exploration of the repertoire of host immune responses to the various parasite stages, and also identification of significant hurdles that need to be overcome. Most important, these advances strengthened the concept that the induction of host immune responses that target all developmental stages of Plasmodium can efficiently control or abrogate Plasmodium infections and strongly support the notion that an effective vaccine can be developed. This vaccine would be a critical component for programs aimed at controlling or eradicating malaria.
Collapse
Affiliation(s)
- Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institute of Health, Rockville, Maryland 20852
| | - Fidel Zavala
- Departmentof Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205
| |
Collapse
|
65
|
Ntege EH, Takashima E, Morita M, Nagaoka H, Ishino T, Tsuboi T. Blood-stage malaria vaccines: post-genome strategies for the identification of novel vaccine candidates. Expert Rev Vaccines 2017; 16:769-779. [PMID: 28604122 DOI: 10.1080/14760584.2017.1341317] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION An efficacious malaria vaccine is necessary to advance the current control measures towards malaria elimination. To-date, only RTS,S/AS01, a leading pre-erythrocytic stage vaccine completed phase 3 trials, but with an efficacy of 28-36% in children, and 18-26% in infants, that waned over time. Blood-stage malaria vaccines protect against disease, and are considered effective targets for the logical design of next generation vaccines to improve the RTS,S field efficacy. Therefore, novel blood-stage vaccine candidate discovery efforts are critical, albeit with several challenges including, high polymorphisms in vaccine antigens, poor understanding of targets of naturally protective immunity, and difficulties in the expression of high AT-rich plasmodial proteins. Areas covered: PubMed ( www.ncbi.nlm.nih.gov/pubmed ) was searched to review the progress and future prospects of malaria vaccine research and development. We focused on post-genome vaccine candidate discovery, malaria vaccine development, sequence diversity, pre-clinical and clinical trials. Expert commentary: Post-genome high-throughput technologies using wheat germ cell-free protein synthesis technology and immuno-profiling with sera from malaria patients with clearly defined outcomes are highlighted to overcome current challenges of malaria vaccine candidate discovery.
Collapse
Affiliation(s)
- Edward H Ntege
- a Division of Malaria Research , Proteo-Science Center, Ehime University , Matsuyama , Ehime , Japan
| | - Eizo Takashima
- a Division of Malaria Research , Proteo-Science Center, Ehime University , Matsuyama , Ehime , Japan
| | - Masayuki Morita
- a Division of Malaria Research , Proteo-Science Center, Ehime University , Matsuyama , Ehime , Japan
| | - Hikaru Nagaoka
- a Division of Malaria Research , Proteo-Science Center, Ehime University , Matsuyama , Ehime , Japan
| | - Tomoko Ishino
- b Division of Molecular Parasitology , Proteo-Science Center, Ehime University , Toon , Ehime , Japan
| | - Takafumi Tsuboi
- a Division of Malaria Research , Proteo-Science Center, Ehime University , Matsuyama , Ehime , Japan
| |
Collapse
|
66
|
Aniweh Y, Gao X, Hao P, Meng W, Lai SK, Gunalan K, Chu TT, Sinha A, Lescar J, Chandramohanadas R, Li HY, Sze SK, Preiser PR. P. falciparum RH5-Basigin interaction induces changes in the cytoskeleton of the host RBC. Cell Microbiol 2017; 19. [PMID: 28409866 DOI: 10.1111/cmi.12747] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 04/06/2017] [Accepted: 04/07/2017] [Indexed: 11/26/2022]
Abstract
The successful invasion of Plasmodium is an essential step in their life cycle. The parasite reticulocyte-binding protein homologues (RHs) and erythrocyte-binding like proteins are two families involved in the invasion leading to merozoite-red blood cell (RBC) junction formation. Ca2+ signaling has been shown to play a critical role in the invasion. RHs have been linked to Ca2+ signaling, which triggers the erythrocyte-binding like proteins release ahead of junction formation, consistent with RHs performing an initial sensing function in identifying suitable RBCs. RH5, the only essential RHs, is a highly promising vaccine candidate. RH5-basigin interaction is essential for merozoite invasion and also important in determining host tropism. Here, we show that RH5 has a distinct function from the other RHs. We show that RH5-Basigin interaction on its own triggers a Ca2+ signal in the RBC resulting in changes in RBC cytoskeletal proteins phosphorylation and overall alterations in RBC cytoskeleton architecture. Antibodies targeting RH5 that block the signal prevent invasion before junction formation consistent with the Ca2+ signal in the RBC leading to rearrangement of the cytoskeleton required for invasion. This work provides the first time a functional context for the essential role of RH5 and will now open up new avenues to target merozoite invasion.
Collapse
Affiliation(s)
- Yaw Aniweh
- Division of Molecular Genetics and Cell biology, Nanyang Technological University, Singapore
| | - Xiaohong Gao
- Division of Molecular Genetics and Cell biology, Nanyang Technological University, Singapore
| | - Piliang Hao
- Division of Structural Biology and Biochemistry, School of Biological Sciences, Nanyang Technological University, Singapore
| | - Wei Meng
- Division of Structural Biology and Biochemistry, School of Biological Sciences, Nanyang Technological University, Singapore
| | - Soak Kuan Lai
- Division of Molecular Genetics and Cell biology, Nanyang Technological University, Singapore
| | - Karthigayan Gunalan
- Division of Molecular Genetics and Cell biology, Nanyang Technological University, Singapore
| | - Trang T Chu
- Pillar of Engineering Product Development, Singapore University of Technology and Design (SUTD), Singapore
| | - Ameya Sinha
- Division of Molecular Genetics and Cell biology, Nanyang Technological University, Singapore.,Pillar of Engineering Product Development, Singapore University of Technology and Design (SUTD), Singapore
| | - Julien Lescar
- Division of Structural Biology and Biochemistry, School of Biological Sciences, Nanyang Technological University, Singapore
| | - Rajesh Chandramohanadas
- Pillar of Engineering Product Development, Singapore University of Technology and Design (SUTD), Singapore
| | - Hoi Yeung Li
- Division of Molecular Genetics and Cell biology, Nanyang Technological University, Singapore
| | - Siu Kwan Sze
- Division of Structural Biology and Biochemistry, School of Biological Sciences, Nanyang Technological University, Singapore
| | - Peter R Preiser
- Division of Molecular Genetics and Cell biology, Nanyang Technological University, Singapore
| |
Collapse
|
67
|
Chen L, Xu Y, Wong W, Thompson JK, Healer J, Goddard-Borger ED, Lawrence MC, Cowman AF. Structural basis for inhibition of erythrocyte invasion by antibodies to Plasmodium falciparum protein CyRPA. eLife 2017; 6. [PMID: 28195530 PMCID: PMC5349848 DOI: 10.7554/elife.21347] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 01/31/2017] [Indexed: 02/01/2023] Open
Abstract
Plasmodium falciparum causes malaria in humans with over 450,000 deaths annually. The asexual blood stage involves invasion of erythrocytes by merozoites, in which they grow and divide to release daughter merozoites, which in turn invade new erythrocytes perpetuating the cycle responsible for malaria. A key step in merozoite invasion is the essential binding of PfRh5/CyRPA/PfRipr complex to basigin, a step linked to the formation of a pore between merozoites and erythrocytes. We show CyRPA interacts directly with PfRh5. An invasion inhibitory monoclonal antibody to CyRPA blocks binding of CyRPA to PfRh5 and complex formation thus illuminating the molecular mechanism for inhibition of parasite growth. We determined the crystal structures of CyRPA alone and in complex with an antibody Fab fragment. CyRPA has a six-bladed β-propeller fold, and we identify the region that interacts with PfRh5. This functionally conserved epitope is a potential target for vaccines against P. falciparum.
Collapse
Affiliation(s)
- Lin Chen
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Yibin Xu
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Wilson Wong
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Jennifer K Thompson
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Julie Healer
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Ethan D Goddard-Borger
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Michael C Lawrence
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Alan F Cowman
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
68
|
Favuzza P, Guffart E, Tamborrini M, Scherer B, Dreyer AM, Rufer AC, Erny J, Hoernschemeyer J, Thoma R, Schmid G, Gsell B, Lamelas A, Benz J, Joseph C, Matile H, Pluschke G, Rudolph MG. Structure of the malaria vaccine candidate antigen CyRPA and its complex with a parasite invasion inhibitory antibody. eLife 2017; 6. [PMID: 28195038 PMCID: PMC5349852 DOI: 10.7554/elife.20383] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 02/06/2017] [Indexed: 12/02/2022] Open
Abstract
Invasion of erythrocytes by Plasmodial merozoites is a composite process involving the interplay of several proteins. Among them, the Plasmodium falciparum Cysteine-Rich Protective Antigen (PfCyRPA) is a crucial component of a ternary complex, including Reticulocyte binding-like Homologous protein 5 (PfRH5) and the RH5-interacting protein (PfRipr), essential for erythrocyte invasion. Here, we present the crystal structures of PfCyRPA and its complex with the antigen-binding fragment of a parasite growth inhibitory antibody. PfCyRPA adopts a 6-bladed β-propeller structure with similarity to the classic sialidase fold, but it has no sialidase activity and fulfills a purely non-enzymatic function. Characterization of the epitope recognized by protective antibodies may facilitate design of peptidomimetics to focus vaccine responses on protective epitopes. Both in vitro and in vivo anti-PfCyRPA and anti-PfRH5 antibodies showed more potent parasite growth inhibitory activity in combination than on their own, supporting a combined delivery of PfCyRPA and PfRH5 in vaccines. DOI:http://dx.doi.org/10.7554/eLife.20383.001 Malaria is one of the deadliest infectious diseases worldwide, killing over 400,000 people a year. About 200 million people are infected every year, placing a huge social and medical burden especially on developing countries. Microscopic parasites known as Plasmodium are responsible for causing this disease. Plasmodium parasites have a complex life cycle involving both mosquito and mammal hosts. This includes a stage where the parasites infect the mammal’s red blood cells, which causes the symptoms of the disease. In 2012, a team of researchers discovered that a protein called CyRPA forms a group (or ‘complex’) with several other proteins to allow the parasites to enter red blood cells. Developing a vaccine is one of the most promising approaches to prevent malaria. Vaccines help the body to recognise and fight an invading microbe by triggering an immune response that results in the production of proteins called antibodies, which can bind to specific molecules on the surface of the microbe. If the microbe later enters the body, these antibodies can be produced quickly to eliminate the microbe before it causes disease. However, efforts to develop a highly effective vaccine against malaria have so far been unsuccessful. Favuzza et al. – including some of the researchers involved in the 2012 work – used a technique called X-ray crystallography to investigate the three-dimensional structure of the CyRPA protein. The experiments show that an antibody is able to bind to a region of CyRPA – a designated ‘protective epitope’ – that is similar in the CyRPA proteins of all Plasmodium falciparum strains. These antibodies can prevent the parasite from entering the red blood cells, and vaccines containing CyRPA may therefore be effective at protecting individuals from malaria. The findings of Favuzza et al. also suggest that using CyRPA in combination with another protein in the complex called RH5 could make the vaccine more powerful as it would make it harder for the parasite to become resistant. The next step following on from this work is to design a vaccine containing protective CyRPA epitopes that triggers an immune response in mammals that is strong enough to reduce the numbers of parasites in the blood. A future challenge will be to develop a vaccine that combines several proteins involved in different stages of the parasite’s life cycle to provide full protection against malaria. DOI:http://dx.doi.org/10.7554/eLife.20383.002
Collapse
Affiliation(s)
- Paola Favuzza
- Medical Parasitology and Infection Biology Department, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Elena Guffart
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Marco Tamborrini
- Medical Parasitology and Infection Biology Department, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Bianca Scherer
- Medical Parasitology and Infection Biology Department, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Anita M Dreyer
- Medical Parasitology and Infection Biology Department, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Arne C Rufer
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Johannes Erny
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Joerg Hoernschemeyer
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Ralf Thoma
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Georg Schmid
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Bernard Gsell
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Araceli Lamelas
- Medical Parasitology and Infection Biology Department, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Joerg Benz
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Catherine Joseph
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Hugues Matile
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Gerd Pluschke
- Medical Parasitology and Infection Biology Department, Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Markus G Rudolph
- Roche Pharmaceutical Research and Early Development, Small Molecule Research, Roche Innovation Center Basel, F Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
69
|
P113 is a merozoite surface protein that binds the N terminus of Plasmodium falciparum RH5. Nat Commun 2017; 8:14333. [PMID: 28186186 PMCID: PMC5309799 DOI: 10.1038/ncomms14333] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 12/16/2016] [Indexed: 01/12/2023] Open
Abstract
Invasion of erythrocytes by Plasmodium falciparum merozoites is necessary for malaria pathogenesis and is therefore a primary target for vaccine development. RH5 is a leading subunit vaccine candidate because anti-RH5 antibodies inhibit parasite growth and the interaction with its erythrocyte receptor basigin is essential for invasion. RH5 is secreted, complexes with other parasite proteins including CyRPA and RIPR, and contains a conserved N-terminal region (RH5Nt) of unknown function that is cleaved from the native protein. Here, we identify P113 as a merozoite surface protein that directly interacts with RH5Nt. Using recombinant proteins and a sensitive protein interaction assay, we establish the binding interdependencies of all the other known RH5 complex components and conclude that the RH5Nt-P113 interaction provides a releasable mechanism for anchoring RH5 to the merozoite surface. We exploit these findings to design a chemically synthesized peptide corresponding to RH5Nt, which could contribute to a cost-effective malaria vaccine. The secreted Plasmodium falciparum protein RH5 is essential for invasion of erythrocytes and is a promising vaccine candidate. Here, Galaway et al. show that the N-terminal region of RH5 binds the GPI-anchored merozoite surface protein P113 and can elicit invasion-blocking antibodies.
Collapse
|
70
|
Abstract
PURPOSE OF REVIEW Red cell receptors provide unique entry points for Plasmodium parasites to initiate blood-stage malaria infection. Parasites encode distinct ligands that bind specifically to both highly abundant and low-copy receptors. Recent advances in the understanding of molecular and structural mechanisms of these interactions provide fundamental insights into receptor-ligand biology and molecular targets for intervention. RECENT FINDINGS The review focuses on the requirements for known interactions, insight derived from complex structures, and mechanisms of receptor/ligand engagement. Further, novel roles for established red cell membrane proteins, parasite ligands and associated interacting partners have recently been established in red cell invasion. SUMMARY The new knowledge underlines the intricacies involved in invasion by a eukaryotic parasite into a eukaryotic host cell demonstrated by expanded parasite ligand families, redundancy in red cell receptor engagement, multitiered temporal binding, and the breadth of receptors engaged.
Collapse
|
71
|
Malaria: Biology and Disease. Cell 2016; 167:610-624. [PMID: 27768886 DOI: 10.1016/j.cell.2016.07.055] [Citation(s) in RCA: 501] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/17/2016] [Accepted: 07/29/2016] [Indexed: 11/22/2022]
Abstract
Malaria has been a major global health problem of humans through history and is a leading cause of death and disease across many tropical and subtropical countries. Over the last fifteen years renewed efforts at control have reduced the prevalence of malaria by over half, raising the prospect that elimination and perhaps eradication may be a long-term possibility. Achievement of this goal requires the development of new tools including novel antimalarial drugs and more efficacious vaccines as well as an increased understanding of the disease and biology of the parasite. This has catalyzed a major effort resulting in development and regulatory approval of the first vaccine against malaria (RTS,S/AS01) as well as identification of novel drug targets and antimalarial compounds, some of which are in human clinical trials.
Collapse
|
72
|
Abstract
Parasites of the genus Plasmodium have a complex life cycle. They alternate between their final mosquito host and their intermediate hosts. The parasite can be either extra- or intracellular, depending on the stage of development. By modifying their shape, motility, and metabolic requirements, the parasite adapts to the different environments in their different hosts. The parasite has evolved to escape the multiple immune mechanisms in the host that try to block parasite development at the different stages of their development. In this article, we describe the mechanisms reported thus far that allow the Plasmodium parasite to evade innate and adaptive immune responses.
Collapse
Affiliation(s)
- Laurent Rénia
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Yun Shan Goh
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore
| |
Collapse
|
73
|
Ntege EH, Arisue N, Ito D, Hasegawa T, Palacpac NM, Egwang TG, Horii T, Takashima E, Tsuboi T. Identification of Plasmodium falciparum reticulocyte binding protein homologue 5-interacting protein, PfRipr, as a highly conserved blood-stage malaria vaccine candidate. Vaccine 2016; 34:5612-5622. [DOI: 10.1016/j.vaccine.2016.09.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/10/2016] [Accepted: 09/15/2016] [Indexed: 10/20/2022]
|
74
|
The association between naturally acquired IgG subclass specific antibodies to the PfRH5 invasion complex and protection from Plasmodium falciparum malaria. Sci Rep 2016; 6:33094. [PMID: 27604417 PMCID: PMC5015043 DOI: 10.1038/srep33094] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/19/2016] [Indexed: 11/13/2022] Open
Abstract
Understanding the targets and mechanisms of human immunity to malaria is important for advancing the development of highly efficacious vaccines and serological tools for malaria surveillance. The PfRH5 and PfRipr proteins form a complex on the surface of P. falciparum merozoites that is essential for invasion of erythrocytes and are vaccine candidates. We determined IgG subclass responses to these proteins among malaria-exposed individuals in Papua New Guinea and their association with protection from malaria in a longitudinal cohort of children. Cytophilic subclasses, IgG1 and IgG3, were predominant with limited IgG2 and IgG4, and IgG subclass-specific responses were higher in older children and those with active infection. High IgG3 to PfRH5 and PfRipr were significantly and strongly associated with reduced risk of malaria after adjusting for potential confounding factors, whereas associations for IgG1 responses were generally weaker and not statistically significant. Results further indicated that malaria exposure leads to the co-acquisition of IgG1 and IgG3 to PfRH5 and PfRipr, as well as to other PfRH invasion ligands, PfRH2 and PfRH4. These findings suggest that IgG3 responses to PfRH5 and PfRipr may play a significant role in mediating naturally-acquired immunity and support their potential as vaccine candidates and their use as antibody biomarkers of immunity.
Collapse
|
75
|
Aniweh Y, Gao X, Gunalan K, Preiser PR. PfRH2b specific monoclonal antibodies inhibit merozoite invasion. Mol Microbiol 2016; 102:386-404. [DOI: 10.1111/mmi.13468] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Yaw Aniweh
- Division of Molecular Genetics and Cell biology, School of Biological Sciences; Nanyang Technological University; 637551 Singapore
| | - Xiaohong Gao
- Division of Molecular Genetics and Cell biology, School of Biological Sciences; Nanyang Technological University; 637551 Singapore
| | - Karthigayan Gunalan
- Division of Molecular Genetics and Cell biology, School of Biological Sciences; Nanyang Technological University; 637551 Singapore
| | - Peter R. Preiser
- Division of Molecular Genetics and Cell biology, School of Biological Sciences; Nanyang Technological University; 637551 Singapore
| |
Collapse
|
76
|
Hjerrild KA, Jin J, Wright KE, Brown RE, Marshall JM, Labbé GM, Silk SE, Cherry CJ, Clemmensen SB, Jørgensen T, Illingworth JJ, Alanine DGW, Milne KH, Ashfield R, de Jongh WA, Douglas AD, Higgins MK, Draper SJ. Production of full-length soluble Plasmodium falciparum RH5 protein vaccine using a Drosophila melanogaster Schneider 2 stable cell line system. Sci Rep 2016; 6:30357. [PMID: 27457156 PMCID: PMC4960544 DOI: 10.1038/srep30357] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/04/2016] [Indexed: 01/27/2023] Open
Abstract
The Plasmodium falciparum reticulocyte-binding protein homolog 5 (PfRH5) has recently emerged as a leading candidate antigen against the blood-stage human malaria parasite. However it has proved challenging to identify a heterologous expression platform that can produce a soluble protein-based vaccine in a manner compliant with current Good Manufacturing Practice (cGMP). Here we report the production of full-length PfRH5 protein using a cGMP-compliant platform called ExpreS(2), based on a Drosophila melanogaster Schneider 2 (S2) stable cell line system. Five sequence variants of PfRH5 were expressed that differed in terms of mutagenesis strategies to remove potential N-linked glycans. All variants bound the PfRH5 receptor basigin and were recognized by a panel of monoclonal antibodies. Analysis following immunization of rabbits identified quantitative and qualitative differences in terms of the functional IgG antibody response against the P. falciparum parasite. The antibodies induced by one protein variant were shown to be qualitatively similar to responses induced by other vaccine platforms. This work identifies Drosophila S2 cells as a clinically-relevant platform suited for the production of 'difficult-to-make' proteins from Plasmodium parasites, and identifies a PfRH5 sequence variant that can be used for clinical production of a non-glycosylated, soluble full-length protein vaccine immunogen.
Collapse
Affiliation(s)
- Kathryn A Hjerrild
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Jing Jin
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Katherine E Wright
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Rebecca E Brown
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Jennifer M Marshall
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Geneviève M Labbé
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Sarah E Silk
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Catherine J Cherry
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Stine B Clemmensen
- ExpreS2ion Biotechnologies, SCION-DTU Science Park, Agern Allé 1, Hørsholm DK-2970, Denmark
| | - Thomas Jørgensen
- ExpreS2ion Biotechnologies, SCION-DTU Science Park, Agern Allé 1, Hørsholm DK-2970, Denmark
| | - Joseph J Illingworth
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Daniel G W Alanine
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Kathryn H Milne
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Rebecca Ashfield
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Willem A de Jongh
- ExpreS2ion Biotechnologies, SCION-DTU Science Park, Agern Allé 1, Hørsholm DK-2970, Denmark
| | - Alexander D Douglas
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Matthew K Higgins
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Simon J Draper
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| |
Collapse
|
77
|
Essential Role of the PfRh5/PfRipr/CyRPA Complex during Plasmodium falciparum Invasion of Erythrocytes. Cell Host Microbe 2016; 20:60-71. [DOI: 10.1016/j.chom.2016.06.004] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 04/05/2016] [Accepted: 05/18/2016] [Indexed: 01/30/2023]
|
78
|
Weiss GE, Crabb BS, Gilson PR. Overlaying Molecular and Temporal Aspects of Malaria Parasite Invasion. Trends Parasitol 2016; 32:284-295. [DOI: 10.1016/j.pt.2015.12.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 12/02/2015] [Accepted: 12/10/2015] [Indexed: 12/31/2022]
|
79
|
Ord RL, Rodriguez M, Lobo CA. Malaria invasion ligand RH5 and its prime candidacy in blood-stage malaria vaccine design. Hum Vaccin Immunother 2016; 11:1465-73. [PMID: 25844685 DOI: 10.1080/21645515.2015.1026496] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
With drug resistance to available therapeutics continuing to develop against Plasmodium falciparum malaria, the development of an effective vaccine candidate remains a major research goal. Successful interruption of invasion of parasites into erythrocytes during the blood stage of infection will prevent the severe clinical symptoms and complications associated with malaria. Previously studied blood stage antigens have highlighted the hurdles that are inherent to this life-cycle stage, namely that highly immunogenic antigens are also globally diverse, resulting in protection only against the vaccine strain, or that naturally acquired immunity to blood stage antigens do not always correlate with actual protection. The blood stage antigen reticulocyte binding homolog RH5 is essential for parasite viability, has globally limited diversity, and is associated with protection from disease. Here we summarize available information on this invasion ligand and recent findings that highlight its candidacy for inclusion in a blood-stage malaria vaccine.
Collapse
Affiliation(s)
- Rosalynn L Ord
- a Blood-Borne Parasites; Lindsley Kimball Research Institute; New York Blood Center ; New York , NY , USA
| | | | | |
Collapse
|
80
|
Abstract
There have been significant decreases in malaria mortality and morbidity in the last 10-15 years, and the most advanced pre-erythrocytic malaria vaccine, RTS,S, received a positive opinion from European regulators in July 2015. However, no blood-stage vaccine has reached a phase III trial. The first part of this review summarizes the pros and cons of various assays and models that have been and will be used to predict the efficacy of blood-stage vaccines. In the second part, blood-stage vaccine candidates that showed some efficacy in human clinical trials or controlled human malaria infection models are discussed. Then, candidates under clinical investigation are described in the third part, and other novel candidates and strategies are reviewed in the last part.
Collapse
Affiliation(s)
- Kazutoyo Miura
- a Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases , National Institutes of Health , Rockville , MD , USA
| |
Collapse
|
81
|
Abstract
PURPOSE OF REVIEW Malaria is caused by the infection and proliferation of parasites from the genus Plasmodium in red blood cells (RBCs). A free Plasmodium parasite, or merozoite, released from an infected RBC must invade another RBC host cell to sustain a blood-stage infection. Here, we review recent advances on RBC invasion by Plasmodium merozoites, focusing on specific molecular interactions between host and parasite. RECENT FINDINGS Recent work highlights the central role of host-parasite interactions at virtually every stage of RBC invasion by merozoites. Biophysical experiments have for the first time measured the strength of merozoite-RBC attachment during invasion. For P. falciparum, there have been many key insights regarding the invasion ligand PfRh5 in particular, including its influence on host species tropism, a co-crystal structure with its RBC receptor basigin, and its suitability as a vaccine target. For P. vivax, researchers identified the origin and emergence of the parasite from Africa, demonstrating a natural link to the Duffy-negative RBC variant in African populations. For the simian parasite P. knowlesi, zoonotic invasion into human cells is linked to RBC age, which has implications for parasitemia during an infection and thus malaria. SUMMARY New studies of the molecular and cellular mechanisms governing RBC invasion by Plasmodium parasites have shed light on various aspects of parasite biology and host cell tropism, and indicate opportunities for malaria control.
Collapse
|
82
|
Beeson JG, Drew DR, Boyle MJ, Feng G, Fowkes FJI, Richards JS. Merozoite surface proteins in red blood cell invasion, immunity and vaccines against malaria. FEMS Microbiol Rev 2016; 40:343-72. [PMID: 26833236 PMCID: PMC4852283 DOI: 10.1093/femsre/fuw001] [Citation(s) in RCA: 243] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2016] [Indexed: 01/11/2023] Open
Abstract
Malaria accounts for an enormous burden of disease globally, with Plasmodium falciparum accounting for the majority of malaria, and P. vivax being a second important cause, especially in Asia, the Americas and the Pacific. During infection with Plasmodium spp., the merozoite form of the parasite invades red blood cells and replicates inside them. It is during the blood-stage of infection that malaria disease occurs and, therefore, understanding merozoite invasion, host immune responses to merozoite surface antigens, and targeting merozoite surface proteins and invasion ligands by novel vaccines and therapeutics have been important areas of research. Merozoite invasion involves multiple interactions and events, and substantial processing of merozoite surface proteins occurs before, during and after invasion. The merozoite surface is highly complex, presenting a multitude of antigens to the immune system. This complexity has proved challenging to our efforts to understand merozoite invasion and malaria immunity, and to developing merozoite antigens as malaria vaccines. In recent years, there has been major progress in this field, and several merozoite surface proteins show strong potential as malaria vaccines. Our current knowledge on this topic is reviewed, highlighting recent advances and research priorities. The authors summarize current knowledge of merozoite surface proteins of malaria parasites; their function in invasion, processing of surface proteins before, during and after invasion, their importance as targets of immunity, and the current status of malaria vaccines that target merozoite surface proteins.
Collapse
Affiliation(s)
- James G Beeson
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, Australia Department of Microbiology, Monash University, Clayton, Victoria, Australia Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Damien R Drew
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, Australia
| | - Michelle J Boyle
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, Australia
| | - Gaoqian Feng
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, Australia
| | - Freya J I Fowkes
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, Australia Department of Epidemiology and Preventive Medicine, Monash University, Clayton, Victoria, Australia School of Population Health, University of Melbourne, Parkville, Victoria, Australia
| | - Jack S Richards
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, Australia Department of Microbiology, Monash University, Clayton, Victoria, Australia Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
83
|
Boes A, Reimann A, Twyman RM, Fischer R, Schillberg S, Spiegel H. A Plant-Based Transient Expression System for the Rapid Production of Malaria Vaccine Candidates. Methods Mol Biol 2016; 1404:597-619. [PMID: 27076325 DOI: 10.1007/978-1-4939-3389-1_39] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
There are currently no vaccines that provide sterile immunity against malaria. Various proteins from different stages of the Plasmodium falciparum life cycle have been evaluated as vaccine candidates, but none of them have fulfilled expectations. Therefore, combinations of key antigens from different stages of the parasites life cycle may be essential for the development of efficacious malaria vaccines. Following the identification of promising antigens using bioinformatics, proteomics, and/or immunological approaches, it is necessary to express, purify, and characterize these proteins and explore the potential of fusion constructs combining different antigens or antigen domains before committing to expensive and time-consuming clinical development. Here, using malaria vaccine candidates as an example, we describe how Agrobacterium tumefaciens-based transient expression in plants can be combined with a modular and flexible cloning strategy as a robust and versatile tool for the rapid production of candidate antigens during research and development.
Collapse
Affiliation(s)
- Alexander Boes
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstrasse 6, 52074, Aachen, Germany
| | - Andreas Reimann
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstrasse 6, 52074, Aachen, Germany
| | | | - Rainer Fischer
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstrasse 6, 52074, Aachen, Germany
- Institute for Molecular Biotechnology, RWTH Aachen University, Worringerweg 1, 52074, Aachen, Germany
| | - Stefan Schillberg
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstrasse 6, 52074, Aachen, Germany
| | - Holger Spiegel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstrasse 6, 52074, Aachen, Germany.
| |
Collapse
|
84
|
Structurally conserved erythrocyte-binding domain in Plasmodium provides a versatile scaffold for alternate receptor engagement. Proc Natl Acad Sci U S A 2015; 113:E191-200. [PMID: 26715754 DOI: 10.1073/pnas.1516512113] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Understanding how malaria parasites gain entry into human red blood cells is essential for developing strategies to stop blood stage infection. Plasmodium vivax preferentially invades reticulocytes, which are immature red blood cells. The organism has two erythrocyte-binding protein families: namely, the Duffy-binding protein (PvDBP) and the reticulocyte-binding protein (PvRBP) families. Several members of the PvRBP family bind reticulocytes, specifically suggesting a role in mediating host cell selectivity of P. vivax. Here, we present, to our knowledge, the first high-resolution crystal structure of an erythrocyte-binding domain from PvRBP2a, solved at 2.12 Å resolution. The monomeric molecule consists of 10 α-helices and one short β-hairpin, and, although the structural fold is similar to that of PfRh5--the essential invasion ligand in Plasmodium falciparum--its surface properties are distinct and provide a possible mechanism for recognition of alternate receptors. Sequence alignments of the crystallized fragment of PvRBP2a with other PvRBPs highlight the conserved placement of disulfide bonds. PvRBP2a binds mature red blood cells through recognition of an erythrocyte receptor that is neuraminidase- and chymotrypsin-resistant but trypsin-sensitive. By examining the patterns of sequence diversity within field isolates, we have identified and mapped polymorphic residues to the PvRBP2a structure. Using mutagenesis, we have also defined the critical residues required for erythrocyte binding. Characterization of the structural features that govern functional erythrocyte binding for the PvRBP family provides a framework for generating new tools that block P. vivax blood stage infection.
Collapse
|
85
|
Hostetler JB, Sharma S, Bartholdson SJ, Wright GJ, Fairhurst RM, Rayner JC. A Library of Plasmodium vivax Recombinant Merozoite Proteins Reveals New Vaccine Candidates and Protein-Protein Interactions. PLoS Negl Trop Dis 2015; 9:e0004264. [PMID: 26701602 PMCID: PMC4689532 DOI: 10.1371/journal.pntd.0004264] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 11/05/2015] [Indexed: 11/27/2022] Open
Abstract
Background A vaccine targeting Plasmodium vivax will be an essential component of any comprehensive malaria elimination program, but major gaps in our understanding of P. vivax biology, including the protein-protein interactions that mediate merozoite invasion of reticulocytes, hinder the search for candidate antigens. Only one ligand-receptor interaction has been identified, that between P. vivax Duffy Binding Protein (PvDBP) and the erythrocyte Duffy Antigen Receptor for Chemokines (DARC), and strain-specific immune responses to PvDBP make it a complex vaccine target. To broaden the repertoire of potential P. vivax merozoite-stage vaccine targets, we exploited a recent breakthrough in expressing full-length ectodomains of Plasmodium proteins in a functionally-active form in mammalian cells and initiated a large-scale study of P. vivax merozoite proteins that are potentially involved in reticulocyte binding and invasion. Methodology/Principal Findings We selected 39 P. vivax proteins that are predicted to localize to the merozoite surface or invasive secretory organelles, some of which show homology to P. falciparum vaccine candidates. Of these, we were able to express 37 full-length protein ectodomains in a mammalian expression system, which has been previously used to express P. falciparum invasion ligands such as PfRH5. To establish whether the expressed proteins were correctly folded, we assessed whether they were recognized by antibodies from Cambodian patients with acute vivax malaria. IgG from these samples showed at least a two-fold change in reactivity over naïve controls in 27 of 34 antigens tested, and the majority showed heat-labile IgG immunoreactivity, suggesting the presence of conformation-sensitive epitopes and native tertiary protein structures. Using a method specifically designed to detect low-affinity, extracellular protein-protein interactions, we confirmed a predicted interaction between P. vivax 6-cysteine proteins P12 and P41, further suggesting that the proteins are natively folded and functional. This screen also identified two novel protein-protein interactions, between P12 and PVX_110945, and between MSP3.10 and MSP7.1, the latter of which was confirmed by surface plasmon resonance. Conclusions/Significance We produced a new library of recombinant full-length P. vivax ectodomains, established that the majority of them contain tertiary structure, and used them to identify predicted and novel protein-protein interactions. As well as identifying new interactions for further biological studies, this library will be useful in identifying P. vivax proteins with vaccine potential, and studying P. vivax malaria pathogenesis and immunity. Trial Registration ClinicalTrials.gov NCT00663546 Plasmodium vivax causes malaria in millions of people each year, primarily in Southeast Asia and Central and South America. P. vivax has a dormant liver stage, which can lead to disease recurrence in infected individuals even in the absence of mosquito transmission. The development of vaccines that target blood-stage P. vivax parasites is therefore likely to be an essential component of any worldwide effort to eradicate malaria. Studying P. vivax is very difficult as this parasite grows poorly in the laboratory and invades only small numbers of young red blood cells in patients. Due to these and other challenges, only a handful of P. vivax proteins have been tested as potential vaccines. To generate more vaccine candidates, we expressed the entire ectodomains of 37 proteins that are predicted to be involved in P. vivax invasion of red blood cells. Antibodies from Cambodian patients with P. vivax malaria recognized heat-sensitive epitopes in the majority of these proteins, suggesting that they are natively folded. We also used the proteins to screen for both predicted and novel protein-protein interactions, confirming that the proteins are functional and further supporting their potential as vaccine candidates. As a new community resource, this P. vivax recombinant protein library will facilitate future studies of P. vivax pathogenesis and immunity, and greatly expands the list of candidate vaccine antigens.
Collapse
Affiliation(s)
- Jessica B. Hostetler
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- Malaria Programme, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Sumana Sharma
- Malaria Programme, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - S. Josefin Bartholdson
- Malaria Programme, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Gavin J. Wright
- Malaria Programme, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Rick M. Fairhurst
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (RMF); (JCR)
| | - Julian C. Rayner
- Malaria Programme, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- * E-mail: (RMF); (JCR)
| |
Collapse
|
86
|
Draper SJ, Angov E, Horii T, Miller LH, Srinivasan P, Theisen M, Biswas S. Recent advances in recombinant protein-based malaria vaccines. Vaccine 2015; 33:7433-43. [PMID: 26458807 PMCID: PMC4687528 DOI: 10.1016/j.vaccine.2015.09.093] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 09/05/2015] [Accepted: 09/28/2015] [Indexed: 01/03/2023]
Abstract
Protein-based vaccines remain the cornerstone approach for B cell and antibody induction against leading target malaria antigens. Advances in antigen selection, immunogen design and epitope-focusing are advancing the field. New heterologous expression platforms are enabling cGMP production of next-generation protein vaccines. Next-generation antigens, protein-based immunogens and virus-like particle (VLP) delivery platforms are in clinical development. Protein-based vaccines will form part of a highly effective multi-component/multi-stage/multi-antigen subunit formulation against malaria.
Plasmodium parasites are the causative agent of human malaria, and the development of a highly effective vaccine against infection, disease and transmission remains a key priority. It is widely established that multiple stages of the parasite's complex lifecycle within the human host and mosquito vector are susceptible to vaccine-induced antibodies. The mainstay approach to antibody induction by subunit vaccination has been the delivery of protein antigen formulated in adjuvant. Extensive efforts have been made in this endeavor with respect to malaria vaccine development, especially with regard to target antigen discovery, protein expression platforms, adjuvant testing, and development of soluble and virus-like particle (VLP) delivery platforms. The breadth of approaches to protein-based vaccines is continuing to expand as innovative new concepts in next-generation subunit design are explored, with the prospects for the development of a highly effective multi-component/multi-stage/multi-antigen formulation seeming ever more likely. This review will focus on recent progress in protein vaccine design, development and/or clinical testing for a number of leading malaria antigens from the sporozoite-, merozoite- and sexual-stages of the parasite's lifecycle–including PfCelTOS, PfMSP1, PfAMA1, PfRH5, PfSERA5, PfGLURP, PfMSP3, Pfs48/45 and Pfs25. Future prospects and challenges for the development, production, human delivery and assessment of protein-based malaria vaccines are discussed.
Collapse
Affiliation(s)
- Simon J Draper
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK.
| | - Evelina Angov
- Walter Reed Army Institute of Research, U. S. Military Malaria Research Program, Malaria Vaccine Branch, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
| | - Toshihiro Horii
- Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 561-873, Japan
| | - Louis H Miller
- Malaria Cell Biology Section, Laboratory of Malaria and Vector Research, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Prakash Srinivasan
- Malaria Cell Biology Section, Laboratory of Malaria and Vector Research, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark; Centre for Medical Parasitology at Department of International Health, Immunology, and Microbiology and Department of Infectious Diseases, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sumi Biswas
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| |
Collapse
|
87
|
Lauron EJ, Aw Yeang HX, Taffner SM, Sehgal RNM. De novo assembly and transcriptome analysis of Plasmodium gallinaceum identifies the Rh5 interacting protein (ripr), and reveals a lack of EBL and RH gene family diversification. Malar J 2015; 14:296. [PMID: 26243218 PMCID: PMC4524024 DOI: 10.1186/s12936-015-0814-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 07/20/2015] [Indexed: 01/01/2023] Open
Abstract
Background Malaria parasites that infect birds can have narrow or broad host-tropisms. These differences in host specificity make avian malaria a useful model for studying the evolution and transmission of parasite assemblages across geographic ranges. The molecular mechanisms involved in host-specificity and the biology of avian malaria parasites in general are important aspects of malaria pathogenesis that warrant further examination. Here, the transcriptome of the malaria parasite Plasmodium gallinaceum was characterized to investigate the biology and the conservation of genes across various malaria parasite species. Methods The P. gallinaceum transcriptome was annotated and KEGG pathway mapping was performed. The ripr gene and orthologous genes that play critical roles in the purine salvage pathway were identified and characterized using bioinformatics and phylogenetic methods. Results Analysis of the transcriptome sequence database identified essential genes of the purine salvage pathway in P. gallinaceum that shared high sequence similarity to Plasmodium falciparum when compared to other mammalian Plasmodium spp. However, based on the current sequence data, there was a lack of orthologous genes that belonged to the erythrocyte-binding-like (EBL) and reticulocyte-binding-like homologue (RH) family in P. gallinaceum. In addition, an orthologue of the Rh5 interacting protein (ripr) was identified. Conclusions These findings suggest that the pathways involved in parasite red blood cell invasion are significantly different in avian Plasmodium parasites, but critical metabolic pathways are conserved throughout divergent Plasmodium taxa. Electronic supplementary material The online version of this article (doi:10.1186/s12936-015-0814-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elvin J Lauron
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
| | - Han Xian Aw Yeang
- Rheumatology Division, Washington University School of Medicine, St. Louis, MO, USA.
| | - Samantha M Taffner
- Rheumatology Division, Washington University School of Medicine, St. Louis, MO, USA.
| | - Ravinder N M Sehgal
- Department of Biology, San Francisco State University, San Francisco, CA, USA.
| |
Collapse
|
88
|
Zenonos ZA, Dummler SK, Müller-Sienerth N, Chen J, Preiser PR, Rayner JC, Wright GJ. Basigin is a druggable target for host-oriented antimalarial interventions. ACTA ACUST UNITED AC 2015. [PMID: 26195724 PMCID: PMC4516795 DOI: 10.1084/jem.20150032] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Zenonos et al. report the development of a new therapeutic for P. falciparum malaria. A recombinant chimeric antibody targeting basigin—a receptor essential for erythrocyte invasion—inhibited parasite invasion and rapidly cleared an established blood-stage infection in vivo. Plasmodium falciparum is the parasite responsible for the most lethal form of malaria, an infectious disease that causes a large proportion of childhood deaths and poses a significant barrier to socioeconomic development in many countries. Although antimalarial drugs exist, the repeated emergence and spread of drug-resistant parasites limit their useful lifespan. An alternative strategy that could limit the evolution of drug-resistant parasites is to target host factors that are essential and universally required for parasite growth. Host-targeted therapeutics have been successfully applied in other infectious diseases but have never been attempted for malaria. Here, we report the development of a recombinant chimeric antibody (Ab-1) against basigin, an erythrocyte receptor necessary for parasite invasion as a putative antimalarial therapeutic. Ab-1 inhibited the PfRH5-basigin interaction and potently blocked erythrocyte invasion by all parasite strains tested. Importantly, Ab-1 rapidly cleared an established P. falciparum blood-stage infection with no overt toxicity in an in vivo infection model. Collectively, our data demonstrate that antibodies or other therapeutics targeting host basigin could be an effective treatment for patients infected with multi-drug resistant P. falciparum.
Collapse
Affiliation(s)
- Zenon A Zenonos
- Cell Surface Signalling Laboratory and Malaria Program, Wellcome Trust Sanger Institute, Cambridge CB10 2DP, England, UK Cell Surface Signalling Laboratory and Malaria Program, Wellcome Trust Sanger Institute, Cambridge CB10 2DP, England, UK
| | - Sara K Dummler
- SMART Singapore-MIT-Alliance for Research and Technology, Infectious Disease IRG, Singapore 138602, Singapore
| | - Nicole Müller-Sienerth
- Cell Surface Signalling Laboratory and Malaria Program, Wellcome Trust Sanger Institute, Cambridge CB10 2DP, England, UK
| | - Jianzhu Chen
- SMART Singapore-MIT-Alliance for Research and Technology, Infectious Disease IRG, Singapore 138602, Singapore Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02420
| | - Peter R Preiser
- SMART Singapore-MIT-Alliance for Research and Technology, Infectious Disease IRG, Singapore 138602, Singapore Nanyang Technological University, School of Biological Sciences, Singapore 637551, Singapore
| | - Julian C Rayner
- Cell Surface Signalling Laboratory and Malaria Program, Wellcome Trust Sanger Institute, Cambridge CB10 2DP, England, UK
| | - Gavin J Wright
- Cell Surface Signalling Laboratory and Malaria Program, Wellcome Trust Sanger Institute, Cambridge CB10 2DP, England, UK Cell Surface Signalling Laboratory and Malaria Program, Wellcome Trust Sanger Institute, Cambridge CB10 2DP, England, UK
| |
Collapse
|
89
|
Boucher LE, Bosch J. The apicomplexan glideosome and adhesins - Structures and function. J Struct Biol 2015; 190:93-114. [PMID: 25764948 PMCID: PMC4417069 DOI: 10.1016/j.jsb.2015.02.008] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Revised: 02/20/2015] [Accepted: 02/26/2015] [Indexed: 01/10/2023]
Abstract
The apicomplexan family of pathogens, which includes Plasmodium spp. and Toxoplasma gondii, are primarily obligate intracellular parasites and invade multiple cell types. These parasites express extracellular membrane protein receptors, adhesins, to form specific pathogen-host cell interaction complexes. Various adhesins are used to invade a variety of cell types. The receptors are linked to an actomyosin motor, which is part of a complex comprised of many proteins known as the invasion machinery or glideosome. To date, reviews on invasion have focused primarily on the molecular pathways and signals of invasion, with little or no structural information presented. Over 75 structures of parasite receptors and glideosome proteins have been deposited with the Protein Data Bank. These structures include adhesins, motor proteins, bridging proteins, inner membrane complex and cytoskeletal proteins, as well as co-crystal structures with peptides and antibodies. These structures provide information regarding key interactions necessary for target receptor engagement, machinery complex formation, how force is transmitted, and the basis of inhibitory antibodies. Additionally, these structures can provide starting points for the development of antibodies and inhibitory molecules targeting protein-protein interactions, with the aim to inhibit invasion. This review provides an overview of the parasite adhesin protein families, the glideosome components, glideosome architecture, and discuss recent work regarding alternative models.
Collapse
Affiliation(s)
- Lauren E Boucher
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA; Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA.
| | - Jürgen Bosch
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA; Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA.
| |
Collapse
|
90
|
Abstract
The development of a highly effective malaria vaccine remains a key goal to aid in the control and eventual eradication of this devastating parasitic disease. The field has made huge strides in recent years, with the first-generation vaccine RTS,S showing modest efficacy in a Phase III clinical trial. The updated 2030 Malaria Vaccine Technology Roadmap calls for a second generation vaccine to achieve 75% efficacy over two years for both Plasmodium falciparum and Plasmodium vivax, and for a vaccine that can prevent malaria transmission. Whole-parasite immunisation approaches and combinations of pre-erythrocytic subunit vaccines are now reporting high-level efficacy, whilst exciting new approaches to the development of blood-stage and transmission-blocking vaccine subunit components are entering clinical development. The development of a highly effective multi-component multi-stage subunit vaccine now appears to be a realistic ambition. This review will cover these recent developments in malaria vaccinology.
Collapse
|
91
|
Weiss GE, Gilson PR, Taechalertpaisarn T, Tham WH, de Jong NWM, Harvey KL, Fowkes FJI, Barlow PN, Rayner JC, Wright GJ, Cowman AF, Crabb BS. Revealing the sequence and resulting cellular morphology of receptor-ligand interactions during Plasmodium falciparum invasion of erythrocytes. PLoS Pathog 2015; 11:e1004670. [PMID: 25723550 PMCID: PMC4344246 DOI: 10.1371/journal.ppat.1004670] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 01/08/2015] [Indexed: 11/18/2022] Open
Abstract
During blood stage Plasmodium falciparum infection, merozoites invade uninfected erythrocytes via a complex, multistep process involving a series of distinct receptor-ligand binding events. Understanding each element in this process increases the potential to block the parasite’s life cycle via drugs or vaccines. To investigate specific receptor-ligand interactions, they were systematically blocked using a combination of genetic deletion, enzymatic receptor cleavage and inhibition of binding via antibodies, peptides and small molecules, and the resulting temporal changes in invasion and morphological effects on erythrocytes were filmed using live cell imaging. Analysis of the videos have shown receptor-ligand interactions occur in the following sequence with the following cellular morphologies; 1) an early heparin-blockable interaction which weakly deforms the erythrocyte, 2) EBA and PfRh ligands which strongly deform the erythrocyte, a process dependant on the merozoite’s actin-myosin motor, 3) a PfRh5-basigin binding step which results in a pore or opening between parasite and host through which it appears small molecules and possibly invasion components can flow and 4) an AMA1–RON2 interaction that mediates tight junction formation, which acts as an anchor point for internalization. In addition to enhancing general knowledge of apicomplexan biology, this work provides a rational basis to combine sequentially acting merozoite vaccine candidates in a single multi-receptor-blocking vaccine. The development of an effective malaria vaccine is a world health priority and would be a critical step toward the control and eventual elimination of this disease. In addition, new pharmacological solutions are necessary as Plasmodium falciparum, the deadliest of the malaria-causing parasites, has developed resistance to every drug currently approved for treatment. Understanding the interactions required for the parasite to invade its erythrocyte host, as well as being valuable to our basic knowledge of parasite biology, is important for the development of drug-based therapies and vaccines. In this study we have, for the first time, filmed P. falciparum parasites invading erythrocytes while systematically blocking several specific interactions between the parasite and the erythrocyte. We have shown there is a sequential progression of specific interactions that occur in at least four distinct steps leading up to invasion. Previous vaccine attempts have targeted one or two of these steps, however, if a single vaccine were designed to block interactions at all four steps, the combined effect might so reduce invasion that parasite growth and disease progression would be arrested. A better understanding of each interaction during invasion, their role and order, can also inform the development of new anti-malarial drugs.
Collapse
Affiliation(s)
| | - Paul R. Gilson
- Burnet Institute, Melbourne, Australia
- Department of Immunology, Monash University, Melbourne, Australia
- * E-mail: (PRG); (BSC)
| | - Tana Taechalertpaisarn
- Burnet Institute, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Australia
| | - Wai-Hong Tham
- Department of Medical Biology, University of Melbourne, Australia
- The Walter & Eliza Hall Institute of Medical Research, Parkville, Australia
| | | | - Katherine L. Harvey
- Burnet Institute, Melbourne, Australia
- Department of Microbiology & Immunology, University of Melbourne, Australia
| | - Freya J. I. Fowkes
- Burnet Institute, Melbourne, Australia
- Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, University of Melbourne, Australia
- Department of Epidemiology and Preventive Medicine and Department of Infectious Diseases, Monash University, Melbourne, Australia
| | - Paul N. Barlow
- Schools of Chemistry and Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Julian C. Rayner
- Malaria Programme, Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Gavin J. Wright
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Alan F. Cowman
- Department of Medical Biology, University of Melbourne, Australia
- The Walter & Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Brendan S. Crabb
- Burnet Institute, Melbourne, Australia
- Department of Immunology, Monash University, Melbourne, Australia
- Department of Microbiology & Immunology, University of Melbourne, Australia
- * E-mail: (PRG); (BSC)
| |
Collapse
|
92
|
PfRH5 as a candidate vaccine for Plasmodium falciparum malaria. Trends Parasitol 2015; 31:87-8. [PMID: 25704640 DOI: 10.1016/j.pt.2015.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 02/06/2015] [Indexed: 11/24/2022]
Abstract
PfRH5 has recently emerged as a strong vaccine candidate for Plasmodium falciparum malaria. Antibodies inhibit invasion of erythrocytes by merozoites and blood-stage replication, and PfRH5 is a significant target of acquired human immunity. Recent studies have shown protective efficacy of PfRH5 vaccines in a non-human primate model of malaria.
Collapse
|
93
|
Conway DJ. Paths to a malaria vaccine illuminated by parasite genomics. Trends Genet 2015; 31:97-107. [PMID: 25620796 PMCID: PMC4359294 DOI: 10.1016/j.tig.2014.12.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 12/19/2014] [Accepted: 12/19/2014] [Indexed: 11/24/2022]
Abstract
Discovery of vaccine candidate antigens by parasite genome sequence analyses. Genetic crosses, linkage group selection, and functional studies on parasites. Characterizing developmental and epigenetic variation alongside allelic polymorphism. Selection by naturally acquired immune responses helps to focus vaccine design.
More human death and disease is caused by malaria parasites than by all other eukaryotic pathogens combined. As early as the sequencing of the first human genome, malaria parasite genomics was prioritized to fuel the discovery of vaccine candidate antigens. This stimulated increased research on malaria, generating new understanding of the cellular and molecular mechanisms of infection and immunity. This review of recent developments illustrates how new approaches in parasite genomics, and increasingly large amounts of data from population studies, are helping to identify antigens that are promising lead targets. Although these results have been encouraging, effective discovery and characterization need to be coupled with more innovation and funding to translate findings into newly designed vaccine products for clinical trials.
Collapse
Affiliation(s)
- David J Conway
- Pathogen Molecular Biology Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK.
| |
Collapse
|
94
|
Multiprotein complex between the GPI-anchored CyRPA with PfRH5 and PfRipr is crucial for Plasmodium falciparum erythrocyte invasion. Proc Natl Acad Sci U S A 2015; 112:1179-84. [PMID: 25583518 DOI: 10.1073/pnas.1415466112] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Erythrocyte invasion by Plasmodium falciparum merozoites is a highly intricate process in which Plasmodium falciparum reticulocyte binding-like homologous protein 5 (PfRH5) is an indispensable parasite ligand that binds with its erythrocyte receptor, Basigin. PfRH5 is a leading blood-stage vaccine candidate because it exhibits limited polymorphisms and elicits potent strain-transcending parasite neutralizing antibodies. However, the mechanism by which it is anchored to the merozoite surface remains unknown because both PfRH5 and the PfRH5-interacting protein (PfRipr) lack transmembrane domains and GPI anchors. Here we have identified a conserved GPI-linked parasite protein, Cysteine-rich protective antigen (CyRPA) as an interacting partner of PfRH5-PfRipr that tethers the PfRH5/PfRipr/CyRPA multiprotein complex on the merozoite surface. CyRPA was demonstrated to be GPI-linked, localized in the micronemes, and essential for erythrocyte invasion. Specific antibodies against the three proteins successfully detected the intact complex in the parasite and coimmunoprecipitated the three interacting partners. Importantly, full-length CyRPA antibodies displayed potent strain-transcending invasion inhibition, as observed for PfRH5. CyRPA does not bind with erythrocytes, suggesting that its parasite neutralizing antibodies likely block its critical interaction with PfRH5-PfRipr, leading to a blockade of erythrocyte invasion. Further, CyRPA and PfRH5 antibody combinations produced synergistic invasion inhibition, suggesting that simultaneous blockade of the PfRH5-Basigin and PfRH5/PfRipr/CyRPA interactions produced an enhanced inhibitory effect. Our discovery of the critical interactions between PfRH5, PfRipr, and the GPI-anchored CyRPA clearly defines the components of the essential PfRH5 adhesion complex for P. falciparum erythrocyte invasion and offers it as a previously unidentified potent target for antimalarial strategies that could abrogate formation of the crucial multiprotein complex.
Collapse
|
95
|
Spiegel H, Boes A, Voepel N, Beiss V, Edgue G, Rademacher T, Sack M, Schillberg S, Reimann A, Fischer R. Application of a Scalable Plant Transient Gene Expression Platform for Malaria Vaccine Development. FRONTIERS IN PLANT SCIENCE 2015; 6:1169. [PMID: 26779197 PMCID: PMC4688378 DOI: 10.3389/fpls.2015.01169] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 12/07/2015] [Indexed: 05/23/2023]
Abstract
Despite decades of intensive research efforts there is currently no vaccine that provides sustained sterile immunity against malaria. In this context, a large number of targets from the different stages of the Plasmodium falciparum life cycle have been evaluated as vaccine candidates. None of these candidates has fulfilled expectations, and as long as we lack a single target that induces strain-transcending protective immune responses, combining key antigens from different life cycle stages seems to be the most promising route toward the development of efficacious malaria vaccines. After the identification of potential targets using approaches such as omics-based technology and reverse immunology, the rapid expression, purification, and characterization of these proteins, as well as the generation and analysis of fusion constructs combining different promising antigens or antigen domains before committing to expensive and time consuming clinical development, represents one of the bottlenecks in the vaccine development pipeline. The production of recombinant proteins by transient gene expression in plants is a robust and versatile alternative to cell-based microbial and eukaryotic production platforms. The transfection of plant tissues and/or whole plants using Agrobacterium tumefaciens offers a low technical entry barrier, low costs, and a high degree of flexibility embedded within a rapid and scalable workflow. Recombinant proteins can easily be targeted to different subcellular compartments according to their physicochemical requirements, including post-translational modifications, to ensure optimal yields of high quality product, and to support simple and economical downstream processing. Here, we demonstrate the use of a plant transient expression platform based on transfection with A. tumefaciens as essential component of a malaria vaccine development workflow involving screens for expression, solubility, and stability using fluorescent fusion proteins. Our results have been implemented for the evidence-based iterative design and expression of vaccine candidates combining suitable P. falciparum antigen domains. The antigens were also produced, purified, and characterized in further studies by taking advantage of the scalability of this platform.
Collapse
Affiliation(s)
- Holger Spiegel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IMEAachen, Germany
| | - Alexander Boes
- Fraunhofer Institute for Molecular Biology and Applied Ecology IMEAachen, Germany
- *Correspondence: Alexander Boes
| | - Nadja Voepel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IMEAachen, Germany
| | - Veronique Beiss
- Fraunhofer Institute for Molecular Biology and Applied Ecology IMEAachen, Germany
| | - Gueven Edgue
- Fraunhofer Institute for Molecular Biology and Applied Ecology IMEAachen, Germany
| | - Thomas Rademacher
- Fraunhofer Institute for Molecular Biology and Applied Ecology IMEAachen, Germany
| | - Markus Sack
- Institute for Molecular Biotechnology, RWTH Aachen UniversityAachen, Germany
| | - Stefan Schillberg
- Fraunhofer Institute for Molecular Biology and Applied Ecology IMEAachen, Germany
| | - Andreas Reimann
- Fraunhofer Institute for Molecular Biology and Applied Ecology IMEAachen, Germany
| | - Rainer Fischer
- Fraunhofer Institute for Molecular Biology and Applied Ecology IMEAachen, Germany
- Institute for Molecular Biotechnology, RWTH Aachen UniversityAachen, Germany
| |
Collapse
|
96
|
Spiegel H, Schinkel H, Kastilan R, Dahm P, Boes A, Scheuermayer M, Chudobová I, Maskus D, Fendel R, Schillberg S, Reimann A, Fischer R. Optimization of a multi-stage, multi-subunit malaria vaccine candidate for the production in Pichia pastoris by the identification and removal of protease cleavage sites. Biotechnol Bioeng 2014; 112:659-67. [PMID: 25335451 DOI: 10.1002/bit.25481] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 09/09/2014] [Accepted: 10/14/2014] [Indexed: 01/21/2023]
Abstract
We demonstrated the successful optimization of a recombinant multi-subunit malaria vaccine candidate protein for production in the methylotrophic yeast Pichia pastoris by the identification and subsequent removal of two protease cleavage sites. After observing protein degradation in the culture supernatant of a fed-batch fermentation, the predominant proteolytic fragment of the secreted recombinant protein was analyzed by mass spectrometry. The MS data indicated the cleavage of an amino acid sequence matching the yeast KEX2-protease consensus motif EKRE. The cleavage in this region was completely abolished by the deletion of the EKRE motif in a modified variant. This modified variant was produced, purified, and used for immunization of rabbits, inducing high antigen specific antibody titers (2 × 10(6) ). Total IgG from rabbit immune sera recognized different stages of Plasmodium falciparum parasites in immunofluorescence assays, indicating native folding of the vaccine candidate. However, the modified variant was still degraded, albeit into different fragments. Further analysis by mass spectrometry and N-terminal sequencing revealed a second cleavage site downstream of the motif PEVK. We therefore removed a 17-amino-acid stretch including the PEVK motif, resulting in the subsequent production of the full-length recombinant vaccine candidate protein without significant degradation, with a yield of 53 mg per liter culture volume. We clearly demonstrate that the proteolytic degradation of recombinant proteins by endogenous P. pastoris proteases can be prevented by the identification and removal of such cleavage sites. This strategy is particularly relevant for the production of recombinant subunit vaccines, where product yield and stability play a more important role than for the production of a stringently-defined native sequence which is necessary for most therapeutic molecules.
Collapse
Affiliation(s)
- Holger Spiegel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstrasse 6, Aachen, 52074, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Wright KE, Hjerrild KA, Bartlett J, Douglas AD, Jin J, Brown RE, Illingworth JJ, Ashfield R, Clemmensen SB, de Jongh WA, Draper SJ, Higgins MK. Structure of malaria invasion protein RH5 with erythrocyte basigin and blocking antibodies. Nature 2014; 515:427-30. [PMID: 25132548 PMCID: PMC4240730 DOI: 10.1038/nature13715] [Citation(s) in RCA: 166] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 07/28/2014] [Indexed: 12/12/2022]
Abstract
Invasion of host erythrocytes is essential to the life cycle of Plasmodium parasites and development of the pathology of malaria. The stages of erythrocyte invasion, including initial contact, apical reorientation, junction formation, and active invagination, are directed by coordinated release of specialized apical organelles and their parasite protein contents. Among these proteins, and central to invasion by all species, are two parasite protein families, the reticulocyte-binding protein homologue (RH) and erythrocyte-binding like proteins, which mediate host-parasite interactions. RH5 from Plasmodium falciparum (PfRH5) is the only member of either family demonstrated to be necessary for erythrocyte invasion in all tested strains, through its interaction with the erythrocyte surface protein basigin (also known as CD147 and EMMPRIN). Antibodies targeting PfRH5 or basigin efficiently block parasite invasion in vitro, making PfRH5 an excellent vaccine candidate. Here we present crystal structures of PfRH5 in complex with basigin and two distinct inhibitory antibodies. PfRH5 adopts a novel fold in which two three-helical bundles come together in a kite-like architecture, presenting binding sites for basigin and inhibitory antibodies at one tip. This provides the first structural insight into erythrocyte binding by the Plasmodium RH protein family and identifies novel inhibitory epitopes to guide design of a new generation of vaccines against the blood-stage parasite.
Collapse
Affiliation(s)
- Katherine E Wright
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Kathryn A Hjerrild
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Jonathan Bartlett
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Alexander D Douglas
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Jing Jin
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Rebecca E Brown
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Joseph J Illingworth
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Rebecca Ashfield
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Stine B Clemmensen
- ExpreS2ion Biotechnologies, SCION-DTU Science Park, Agern Allé 1, DK-2970 Horsholm, Denmark
| | - Willem A de Jongh
- ExpreS2ion Biotechnologies, SCION-DTU Science Park, Agern Allé 1, DK-2970 Horsholm, Denmark
| | - Simon J Draper
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Matthew K Higgins
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| |
Collapse
|
98
|
Chen L, Xu Y, Healer J, Thompson JK, Smith BJ, Lawrence MC, Cowman AF. Crystal structure of PfRh5, an essential P. falciparum ligand for invasion of human erythrocytes. eLife 2014; 3. [PMID: 25296023 PMCID: PMC4356141 DOI: 10.7554/elife.04187] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 10/07/2014] [Indexed: 01/15/2023] Open
Abstract
Plasmodium falciparum causes the most severe form of malaria in humans and is responsible for over 700,000 deaths annually. It is an obligate intracellular parasite and invades erythrocytes where it grows in a relatively protected niche. Invasion of erythrocytes is essential for parasite survival and this involves interplay of multiple protein–protein interactions. One of the most important interactions is binding of parasite invasion ligand families EBLs and PfRhs to host receptors on the surface of erythrocytes. PfRh5 is the only essential invasion ligand within the PfRh family and is an important vaccine candidate. PfRh5 binds the host receptor basigin. In this study, we have determined the crystal structure of PfRh5 using diffraction data to 2.18 Å resolution. PfRh5 exhibits a novel fold, comprising nine mostly anti-parallel α-helices encasing an N-terminal β-hairpin, with the overall shape being an elliptical disk. This is the first three-dimensional structure determined for the PfRh family of proteins.
Collapse
Affiliation(s)
- Lin Chen
- Division of Infection and Immunity, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Yibin Xu
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Julie Healer
- Division of Infection and Immunity, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Jenny K Thompson
- Division of Infection and Immunity, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Brian J Smith
- Department of Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Michael C Lawrence
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Alan F Cowman
- Division of Infection and Immunity, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| |
Collapse
|
99
|
Chiu CYH, Healer J, Thompson JK, Chen L, Kaul A, Savergave L, Raghuwanshi A, Li Wai Suen CSN, Siba PM, Schofield L, Mueller I, Cowman AF, Hansen DS. Association of antibodies to Plasmodium falciparum reticulocyte binding protein homolog 5 with protection from clinical malaria. Front Microbiol 2014; 5:314. [PMID: 25071730 PMCID: PMC4074990 DOI: 10.3389/fmicb.2014.00314] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 06/09/2014] [Indexed: 11/13/2022] Open
Abstract
Emerging evidence suggests that antibodies against merozoite proteins involved in Plasmodium falciparum invasion into the red blood cell (RBC) play an important role in clinical immunity to malaria. The protein family of parasite antigens known as P. falciparum reticulocyte binding protein-like homolog (PfRh) is required for RBC invasion. PfRh5 is the only member within the PfRh family that cannot be genetically deleted, suggesting it plays an essential role in parasite survival. This antigen forms a complex with the cysteine-rich P. falciparum Rh5 interacting protein (PfRipr), on the merozoite surface during RBC invasion. The PfRh5 ectodomain sequence and a C-terminal fragment of PfRipr were cloned and expressed in Escherichia coli and baculovirus-infected cells, respectively. Immunization of rabbits with these recombinant proteins induced antibodies able to inhibit growth of various P. falciparum strains. Antibody responses to these proteins were investigated in a treatment-re-infection study conducted in an endemic area of Papua New Guinea (PNG) to determine their contribution to naturally acquired immunity. Antibody titers to PfRh5 but not PfRipr showed strong association with protection against P. falciparum clinical episodes. When associations with time-to-first infection were analyzed, high antibody levels against PfRh5 were also found to be associated with protection from high-density infections but not from re-infection. Together these results indicate that PfRh5 is an important target of protective immunity and constitutes a promising vaccine candidate.
Collapse
Affiliation(s)
- Chris Y H Chiu
- The Walter and Eliza Hall Institute of Medical Research Parkville, VIC, Australia ; Department of Medical Biology, The University of Melbourne Parkville, VIC, Australia
| | - Julie Healer
- The Walter and Eliza Hall Institute of Medical Research Parkville, VIC, Australia
| | - Jennifer K Thompson
- The Walter and Eliza Hall Institute of Medical Research Parkville, VIC, Australia
| | - Lin Chen
- The Walter and Eliza Hall Institute of Medical Research Parkville, VIC, Australia
| | - Aiki Kaul
- Gennova Biopharmaceuticals Pune, India
| | | | | | | | - Peter M Siba
- Vector Borne Disease Unit, Papua New Guinea Institute of Medical Research Goroka, New Guinea
| | - Louis Schofield
- The Walter and Eliza Hall Institute of Medical Research Parkville, VIC, Australia ; Australian Institute of Tropical Health and Medicine, James Cook University Douglas, QLD, Australia
| | - Ivo Mueller
- The Walter and Eliza Hall Institute of Medical Research Parkville, VIC, Australia ; Barcelona Center for International Health, University of Barcelona Barcelona, Spain
| | - Alan F Cowman
- The Walter and Eliza Hall Institute of Medical Research Parkville, VIC, Australia ; Department of Medical Biology, The University of Melbourne Parkville, VIC, Australia
| | - Diana S Hansen
- The Walter and Eliza Hall Institute of Medical Research Parkville, VIC, Australia ; Department of Medical Biology, The University of Melbourne Parkville, VIC, Australia
| |
Collapse
|
100
|
Affiliation(s)
- Gavin J. Wright
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
- Malaria Programme, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
- * E-mail: (GJW); (JCR)
| | - Julian C. Rayner
- Malaria Programme, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
- * E-mail: (GJW); (JCR)
| |
Collapse
|