1
|
Langowski MD, Francica JR, Roederer AL, Hurlburt NK, Rodarte JV, Da Silva Pereira L, Flynn BJ, Bonilla B, Dillon M, Kiyuka P, Ravichandran R, Weidle C, Carter L, Rao M, Matyas GR, Pepper M, Idris AH, Seder RA, Pancera M, King NP. Elicitation of liver-stage immunity by nanoparticle immunogens displaying P. falciparum CSP-derived antigens. NPJ Vaccines 2025; 10:87. [PMID: 40325041 PMCID: PMC12053698 DOI: 10.1038/s41541-025-01140-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 04/23/2025] [Indexed: 05/07/2025] Open
Abstract
A vaccine that provides robust, durable protection against malaria remains a global health priority. Although a breakthrough in the fight against malaria has recently been achieved by the licensure of two vaccines based on the circumsporozoite protein (CSP), the effectiveness and durability of protection can still be improved. Both vaccines contain a portion of CSP that does not include epitopes targeted by recently identified, potently protective monoclonal antibodies, suggesting that newer immunogens can expand the breadth of immunity and potentially increase protection. Here we explored >100 alternative CSP-based immunogens and evaluated the immunogenicity and protection of a large number of candidates, comparing several to the licensed R21 vaccine. The data highlight several general features that improve the stability and immunogenicity of CSP-based vaccines, such as inclusion of the C-terminal domain and high-density display on protein nanoparticle scaffolds. We also identify antigen design strategies that do not warrant further exploration, such as synthetic repeat regions that include non-native repeat cadences. The benchmark R21 vaccine outperformed our best immunogen for immunogenicity and protection. Overall, our data provide valuable insights on the inclusion of junctional region epitopes that will guide the development of potent and durable vaccines against malaria.
Collapse
Affiliation(s)
- Mark D Langowski
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, WA, USA
| | - Joseph R Francica
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alex L Roederer
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Nicholas K Hurlburt
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Justas V Rodarte
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lais Da Silva Pereira
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Barbara J Flynn
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Brian Bonilla
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Marlon Dillon
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Patience Kiyuka
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rashmi Ravichandran
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Connor Weidle
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lauren Carter
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Mangala Rao
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- U.S. Military HIV Research Program, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Gary R Matyas
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- U.S. Military HIV Research Program, Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Marion Pepper
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Azza H Idris
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Marie Pancera
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| | - Neil P King
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
| |
Collapse
|
2
|
Baptista SJS, Lahree A, Marques S, Bento I, Mello-Vieira J, Mendes AM, Zuzarte-Luís V, Mota MM. CSP ubiquitylation favours Plasmodium berghei survival during early liver stage infection. Sci Rep 2025; 15:14498. [PMID: 40281042 PMCID: PMC12032137 DOI: 10.1038/s41598-025-98294-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
The circumsporozoite protein (CSP), an essential protein that covers the surface of the Plasmodium sporozoite, is a key player in multiple stages of the parasite development within the mosquito and during interactions between sporozoites and mammalian hepatocytes. Here, we identify a novel function of Plasmodium berghei CSP: preventing parasite elimination during the early stages of hepatic infection, through its ubiquitylation at two lysine (K) residues, K252 and K258, located in the C-terminal domain. A Plasmodium berghei transgenic line lacking these lysine residues exhibited a significant decrease in hepatic infectivity, with parasites being eliminated 4 h after infection. The reduced infectivity correlated with an increased association of host autophagy markers, LC3 and LAMP1, to the parasitophorous vacuole membrane of the liver stage parasite. Notably, inhibiting the host autophagy pathway fully rescued the mutant parasites from elimination. Collectively, we reveal a strategy employed by Plasmodium to evade early clearance during hepatic infection, which relies on the ubiquitylation of specific CSP lysine residues, that results in reduced parasite elimination via host autophagic and lysosomal activity.
Collapse
Affiliation(s)
- Sara J S Baptista
- Gulbenkian Institute for Molecular Medicine, 1649-028, Lisbon, Portugal
- Instituto de Medicina Molecular JLA, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - Aparajita Lahree
- Instituto de Medicina Molecular JLA, Universidade de Lisboa, 1649-028, Lisbon, Portugal
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, Dresden, Germany
| | - Sofia Marques
- Gulbenkian Institute for Molecular Medicine, 1649-028, Lisbon, Portugal
- Instituto de Medicina Molecular JLA, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - Inês Bento
- Gulbenkian Institute for Molecular Medicine, 1649-028, Lisbon, Portugal
- Instituto de Medicina Molecular JLA, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - João Mello-Vieira
- Instituto de Medicina Molecular JLA, Universidade de Lisboa, 1649-028, Lisbon, Portugal
- Faculty of Medicine, Institute of Biochemistry 2 and Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - António M Mendes
- Instituto de Medicina Molecular JLA, Universidade de Lisboa, 1649-028, Lisbon, Portugal
- SGS Portugal S.A., Polo Tecnológico de Lisboa, R. Cesina Adães Bermudes Lote 11 N° 1, 1600-604, Lisbon, Portugal
| | - Vanessa Zuzarte-Luís
- Instituto de Medicina Molecular JLA, Universidade de Lisboa, 1649-028, Lisbon, Portugal
- SGS Portugal S.A., Polo Tecnológico de Lisboa, R. Cesina Adães Bermudes Lote 11 N° 1, 1600-604, Lisbon, Portugal
| | - Maria M Mota
- Gulbenkian Institute for Molecular Medicine, 1649-028, Lisbon, Portugal.
- Instituto de Medicina Molecular JLA, Universidade de Lisboa, 1649-028, Lisbon, Portugal.
| |
Collapse
|
3
|
Zanghí G, Patel H, Smith JL, Camargo N, Bae Y, Hesping E, Boddey JA, Venugopal K, Marti M, Flannery EL, Chuenchob V, Fishbaugher ME, Mikolajczak SA, Roobsoong W, Sattabongkot J, Gupta P, Pazzagli L, Rezakhani N, Betz W, Hayes K, Goswami D, Vaughan AM, Kappe SHI. Genome-wide gene expression profiles throughout human malaria parasite liver stage development in humanized mice. Nat Microbiol 2025; 10:569-584. [PMID: 39891010 PMCID: PMC11790487 DOI: 10.1038/s41564-024-01905-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 12/04/2024] [Indexed: 02/03/2025]
Abstract
Gene expression of Plasmodium falciparum (Pf) liver-stage (LS) parasites has remained poorly characterized, although they are major vaccine and drug targets. Using a human liver-chimaeric mouse model and a fluorescent parasite line (PfNF54CSPGFP), we isolated PfLS and performed transcriptomics on key LS developmental phases. We linked clustered gene expression to ApiAP2, a major family of transcription factors that regulate the parasite life cycle. This provided insights into transcriptional regulation of LS infection and expression of essential LS metabolic and biosynthetic pathways. We observed expression of antigenically variant PfEMP1 proteins and the major Pf protein export machine PTEX and identified protein candidates that might be exported by LS parasites. Comparing Pf and P. vivax LS transcriptomes, we uncovered differences in their expression of sexual commitment factors. This data will aid LS research and vaccine and drug target identification for prevention of malaria infection.
Collapse
Affiliation(s)
- Gigliola Zanghí
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA.
| | - Hardik Patel
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Jenny L Smith
- Research Scientific Computing, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nelly Camargo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Yeji Bae
- Research Scientific Computing, Seattle Children's Research Institute, Seattle, WA, USA
| | - Eva Hesping
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Justin A Boddey
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Kannan Venugopal
- Institute for Parasitology, University of Zurich, Zurich, Switzerland
- Institute of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Matthias Marti
- Institute for Parasitology, University of Zurich, Zurich, Switzerland
- Institute of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Erika L Flannery
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
- Global Health, Biomedical Research, Novartis, Emeryville, CA, USA
| | - Vorada Chuenchob
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
- Global Health, Biomedical Research, Novartis, Emeryville, CA, USA
| | - Matthew E Fishbaugher
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
- Global Health, Biomedical Research, Novartis, Emeryville, CA, USA
| | - Sebastian A Mikolajczak
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Priya Gupta
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Lucia Pazzagli
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nastaran Rezakhani
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - William Betz
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Kiera Hayes
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Debashree Goswami
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Ashley M Vaughan
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| |
Collapse
|
4
|
Schmid M, Beyeler R, Caldelari R, Rehmann R, Heussler V, Roques M. Generation of a genetically double-attenuated Plasmodium berghei parasite that fully arrests growth during late liver stage development. PLoS One 2024; 19:e0316164. [PMID: 39739824 DOI: 10.1371/journal.pone.0316164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/06/2024] [Indexed: 01/02/2025] Open
Abstract
Malaria caused by Plasmodium parasites remains a large health burden. One approach to combat this disease involves vaccinating individuals with whole sporozoites that have been genetically modified to arrest their development at a specific stage in the liver by targeted gene deletion, resulting in a genetically attenuated parasite (GAP). Through a comprehensive phenotyping screen, we identified the hscb gene, encoding a putative iron-sulfur protein assembly chaperone, as crucial for liver stage development, making it a suitable candidate gene for GAP generation. Parasites lacking Plasmodium berghei HscB (PbHscB) exhibited normal sporozoite production in mosquitoes, but their liver stage development was severely impaired, characterized by slow growth and delayed expression of merozoite surface protein 1 (MSP1). In vivo experiments demonstrated that PbHscB-deficient parasites exhibited a delay in prepatency of 2-4 days, emphasizing the significance of PbHscB for exo-erythrocytic development. Although knockout of PbHscB alone allowed breakthrough infections, it is a potent candidate for a dual gene deletion strategy. PlasMei2, an RNA-binding protein, was previously found to be crucial for the completion of liver stage development. We generated a PbHscB-PbMei2-double attenuated parasite line, serving as a late liver stage-arresting replication-competent (LARC) GAP, providing a solid block of liver-to-blood stage transition.
Collapse
Affiliation(s)
- Melanie Schmid
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Raphael Beyeler
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Reto Caldelari
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Ruth Rehmann
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Volker Heussler
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Magali Roques
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| |
Collapse
|
5
|
Singer M, Kanatani S, Castillo SG, Frischknecht F, Sinnis P. The Plasmodium circumsporozoite protein. Trends Parasitol 2024; 40:1124-1134. [PMID: 39572325 DOI: 10.1016/j.pt.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 12/07/2024]
Abstract
The circumsporozoite protein (CSP) is one of the most studied proteins of the malaria parasite. It is the target of the only licensed malaria vaccines and is essential for sporozoite formation and infectivity. Yet, the mechanisms by which CSP functions and its interactions with other proteins are only beginning to be understood. Here we review the current state of knowledge of CSP structure and function, as sporozoites develop in the mosquito and establish infection in the mammalian host, and outline outstanding questions that need to be addressed.
Collapse
Affiliation(s)
- Mirko Singer
- Parasitology, Center for Infectious Diseases, Heidelberg University Medical Faculty, Heidelberg, Germany
| | - Sachie Kanatani
- Johns Hopkins School of Public Health and Johns Hopkins Malaria Research Institute, 615 North Wolfe Street, Baltimore, MD, USA
| | - Stefano Garcia Castillo
- Johns Hopkins School of Public Health and Johns Hopkins Malaria Research Institute, 615 North Wolfe Street, Baltimore, MD, USA
| | - Friedrich Frischknecht
- Parasitology, Center for Infectious Diseases, Heidelberg University Medical Faculty, Heidelberg, Germany; German Center for Infection Research, partner site Heidelberg, Heidelberg, Germany
| | - Photini Sinnis
- Johns Hopkins School of Public Health and Johns Hopkins Malaria Research Institute, 615 North Wolfe Street, Baltimore, MD, USA.
| |
Collapse
|
6
|
McConville R, Krol JMM, Steel RWJ, O’Neill MT, Davey BK, Hodder AN, Nebl T, Cowman AF, Kneteman N, Boddey JA. Flp/ FRT-mediated disruption of ptex150 and exp2 in Plasmodium falciparum sporozoites inhibits liver-stage development. Proc Natl Acad Sci U S A 2024; 121:e2403442121. [PMID: 38968107 PMCID: PMC11252984 DOI: 10.1073/pnas.2403442121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/31/2024] [Indexed: 07/07/2024] Open
Abstract
Plasmodium falciparum causes severe malaria and assembles a protein translocon (PTEX) complex at the parasitophorous vacuole membrane (PVM) of infected erythrocytes, through which several hundred proteins are exported to facilitate growth. The preceding liver stage of infection involves growth in a hepatocyte-derived PVM; however, the importance of protein export during P. falciparum liver infection remains unexplored. Here, we use the FlpL/FRT system to conditionally excise genes in P. falciparum sporozoites for functional liver-stage studies. Disruption of PTEX members ptex150 and exp2 did not affect sporozoite development in mosquitoes or infectivity for hepatocytes but attenuated liver-stage growth in humanized mice. While PTEX150 deficiency reduced fitness on day 6 postinfection by 40%, EXP2 deficiency caused 100% loss of liver parasites, demonstrating that PTEX components are required for growth in hepatocytes to differing degrees. To characterize PTEX loss-of-function mutations, we localized four liver-stage Plasmodium export element (PEXEL) proteins. P. falciparum liver specific protein 2 (LISP2), liver-stage antigen 3 (LSA3), circumsporozoite protein (CSP), and a Plasmodium berghei LISP2 reporter all localized to the periphery of P. falciparum liver stages but were not exported beyond the PVM. Expression of LISP2 and CSP but not LSA3 was reduced in ptex150-FRT and exp2-FRT liver stages, suggesting that expression of some PEXEL proteins is affected directly or indirectly by PTEX disruption. These results show that PTEX150 and EXP2 are important for P. falciparum development in hepatocytes and emphasize the emerging complexity of PEXEL protein trafficking.
Collapse
Affiliation(s)
- Robyn McConville
- Division of Infectious Diseases & Immune Defence, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC3010, Australia
| | - Jelte M. M. Krol
- Division of Infectious Diseases & Immune Defence, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC3010, Australia
| | - Ryan W. J. Steel
- Division of Infectious Diseases & Immune Defence, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC3010, Australia
| | - Matthew T. O’Neill
- Division of Infectious Diseases & Immune Defence, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC3052, Australia
| | - Bethany K. Davey
- Division of Infectious Diseases & Immune Defence, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC3010, Australia
| | - Anthony N. Hodder
- Division of Infectious Diseases & Immune Defence, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC3010, Australia
| | - Thomas Nebl
- Division of Infectious Diseases & Immune Defence, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC3010, Australia
| | - Alan F. Cowman
- Division of Infectious Diseases & Immune Defence, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC3010, Australia
| | - Norman Kneteman
- Departments of Surgery, University of Alberta, Edmonton, ABT6G 2E1, Canada
| | - Justin A. Boddey
- Division of Infectious Diseases & Immune Defence, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC3010, Australia
| |
Collapse
|
7
|
Haskins BE, Gullicksrud JA, Wallbank BA, Dumaine JE, Guérin A, Cohn IS, O'Dea KM, Pardy RD, Merolle MI, Shallberg LA, Hunter EN, Byerly JH, Smith EJ, Buenconsejo GY, McLeod BI, Christian DA, Striepen B, Hunter CA. Dendritic cell-mediated responses to secreted Cryptosporidium effectors promote parasite-specific CD8 + T cell responses. Mucosal Immunol 2024; 17:387-401. [PMID: 38508522 PMCID: PMC11193387 DOI: 10.1016/j.mucimm.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 03/22/2024]
Abstract
Cryptosporidium causes debilitating diarrheal disease in patients with primary and acquired defects in T cell function. However, it has been a challenge to understand how this infection generates T cell responses and how they mediate parasite control. Here, Cryptosporidium was engineered to express a parasite effector protein (MEDLE-2) that contains the major histocompatibility complex-I restricted SIINFEKL epitope which is recognized by T cell receptor transgenic OT-I(OVA-TCR-I) clusters of differentiation (CD)8+ T cells. These modified parasites induced expansion of endogenous SIINFEKL-specific and OT-I CD8+ T cells that were a source of interferon-gamma (IFN-γ) that could restrict growth of Cryptosporidium. This T cell response was dependent on the translocation of the effector and similar results were observed with another secreted parasite effector (rhoptry protein 1). Although infection and these translocated effector proteins are restricted to intestinal epithelial cells, type 1 conventional dendritic cells were required to generate CD8+ T cell responses to these model antigens. These data sets highlight Cryptosporidium effectors as potential targets of the immune system and suggest that crosstalk between enterocytes and type 1 conventional dendritic cells is crucial for CD8+ T cell responses to Cryptosporidium.
Collapse
Affiliation(s)
- Breanne E Haskins
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Jodi A Gullicksrud
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA; Cell Press, Cambridge, Massachusetts, USA
| | - Bethan A Wallbank
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Jennifer E Dumaine
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Amandine Guérin
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Ian S Cohn
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Keenan M O'Dea
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Ryan D Pardy
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Maria I Merolle
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Lindsey A Shallberg
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Emma N Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Jessica H Byerly
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Eleanor J Smith
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Gracyn Y Buenconsejo
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Briana I McLeod
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - David A Christian
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Boris Striepen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, USA.
| |
Collapse
|
8
|
Shrikondawar AN, Chennoju K, Ghosh DK, Ranjan A. Identification and characterization of nuclear localization signals in the circumsporozoite protein of Plasmodium falciparum. FEBS Lett 2024; 598:801-817. [PMID: 38369616 DOI: 10.1002/1873-3468.14829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/30/2023] [Accepted: 01/15/2024] [Indexed: 02/20/2024]
Abstract
Secretory proteins of Plasmodium exhibit differential spatial and functional activity within the host cell nucleus. However, the nuclear localization signals (NLSs) for these proteins remain largely uncharacterized. In this study, we have identified and characterized two NLSs in the circumsporozoite protein of Plasmodium falciparum (Pf-CSP). Both NLSs in the Pf-CSP contain clusters of lysine and arginine residues essential for specific interactions with the conserved tryptophan and asparagine residues of importin-α, facilitating nuclear translocation of Pf-CSP. While the two NLSs of Pf-CSP function independently and are both crucial for nuclear localization, a single NLS of Pf-CSP leads to weak nuclear localization. These findings shed light on the mechanism of nuclear penetrability of secretory proteins of Plasmodium proteins.
Collapse
Affiliation(s)
- Akshaykumar Nanaji Shrikondawar
- Computational and Functional Genomics Group, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, India
- Graduate Studies, Regional Centre for Biotechnology, Faridabad, India
| | - Kiranmai Chennoju
- Computational and Functional Genomics Group, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, India
- Graduate Studies, Manipal Academy of Higher Education, Manipal, India
| | | | - Akash Ranjan
- Computational and Functional Genomics Group, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, India
| |
Collapse
|
9
|
Zhu C, Jiao S, Xu W. CD8 + Trms against malaria liver-stage: prospects and challenges. Front Immunol 2024; 15:1344941. [PMID: 38318178 PMCID: PMC10839007 DOI: 10.3389/fimmu.2024.1344941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Attenuated sporozoites provide a valuable model for exploring protective immunity against the malarial liver stage, guiding the design of highly efficient vaccines to prevent malaria infection. Liver tissue-resident CD8+ T cells (CD8+ Trm cells) are considered the host front-line defense against malaria and are crucial to developing prime-trap/target strategies for pre-erythrocytic stage vaccine immunization. However, the spatiotemporal regulatory mechanism of the generation of liver CD8+ Trm cells and their responses to sporozoite challenge, as well as the protective antigens they recognize remain largely unknown. Here, we discuss the knowledge gap regarding liver CD8+ Trm cell formation and the potential strategies to identify predominant protective antigens expressed in the exoerythrocytic stage, which is essential for high-efficacy malaria subunit pre-erythrocytic vaccine designation.
Collapse
Affiliation(s)
- Chengyu Zhu
- The School of Medicine, Chongqing University, Chongqing, China
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shiming Jiao
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wenyue Xu
- The School of Medicine, Chongqing University, Chongqing, China
- Department of Pathogenic Biology, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
10
|
Ganley M, Holz LE, Minnell JJ, de Menezes MN, Burn OK, Poa KCY, Draper SL, English K, Chan STS, Anderson RJ, Compton BJ, Marshall AJ, Cozijnsen A, Chua YC, Ge Z, Farrand KJ, Mamum JC, Xu C, Cockburn IA, Yui K, Bertolino P, Gras S, Le Nours J, Rossjohn J, Fernandez-Ruiz D, McFadden GI, Ackerley DF, Painter GF, Hermans IF, Heath WR. mRNA vaccine against malaria tailored for liver-resident memory T cells. Nat Immunol 2023; 24:1487-1498. [PMID: 37474653 DOI: 10.1038/s41590-023-01562-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 06/15/2023] [Indexed: 07/22/2023]
Abstract
Malaria is caused by Plasmodium species transmitted by Anopheles mosquitoes. Following a mosquito bite, Plasmodium sporozoites migrate from skin to liver, where extensive replication occurs, emerging later as merozoites that can infect red blood cells and cause symptoms of disease. As liver tissue-resident memory T cells (Trm cells) have recently been shown to control liver-stage infections, we embarked on a messenger RNA (mRNA)-based vaccine strategy to induce liver Trm cells to prevent malaria. Although a standard mRNA vaccine was unable to generate liver Trm or protect against challenge with Plasmodium berghei sporozoites in mice, addition of an agonist that recruits T cell help from type I natural killer T cells under mRNA-vaccination conditions resulted in significant generation of liver Trm cells and effective protection. Moreover, whereas previous exposure of mice to blood-stage infection impaired traditional vaccines based on attenuated sporozoites, mRNA vaccination was unaffected, underlining the potential for such a rational mRNA-based strategy in malaria-endemic regions.
Collapse
Affiliation(s)
- Mitch Ganley
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Lauren E Holz
- Department of Microbiology and Immunology, The Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | | | - Maria N de Menezes
- Department of Microbiology and Immunology, The Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | - Olivia K Burn
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Kean Chan Yew Poa
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Sarah L Draper
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Kieran English
- Centenary Institute and University of Sydney, AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Susanna T S Chan
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Regan J Anderson
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Benjamin J Compton
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Andrew J Marshall
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand
| | - Anton Cozijnsen
- School of BioSciences, University of Melbourne, Parkville, Victoria, Australia
| | - Yu Cheng Chua
- Department of Microbiology and Immunology, The Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | - Zhengyu Ge
- Department of Microbiology and Immunology, The Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | | | - John C Mamum
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Calvin Xu
- Department of Microbiology and Immunology, The Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | - Ian A Cockburn
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Katsuyuki Yui
- Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Sakamoto, Nagasaki, Japan
| | - Patrick Bertolino
- Centenary Institute and University of Sydney, AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Stephanie Gras
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Jérôme Le Nours
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - Daniel Fernandez-Ruiz
- Department of Microbiology and Immunology, The Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | - Geoffrey I McFadden
- School of BioSciences, University of Melbourne, Parkville, Victoria, Australia
| | - David F Ackerley
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Gavin F Painter
- Ferrier Research Institute, Victoria University of Wellington, Lower Hutt, New Zealand.
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand.
| | - Ian F Hermans
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand.
- Malaghan Institute of Medical Research, Wellington, New Zealand.
| | - William R Heath
- Department of Microbiology and Immunology, The Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
11
|
Haskins BE, Gullicksrud JA, Wallbank BA, Dumaine JE, Guérin A, Cohn IS, O'Dea KM, Pardy RD, Merolle MI, Shallberg LA, Hunter EN, Byerly JH, Smith EJ, Buenconsejo GY, McLeod BI, Christian DA, Striepen B, Hunter CA. Dendritic cell-mediated responses to secreted Cryptosporidium effectors are required for parasite-specific CD8 + T cell responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.16.553566. [PMID: 37645924 PMCID: PMC10462095 DOI: 10.1101/2023.08.16.553566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Cryptosporidium causes debilitating diarrheal disease in patients with primary and acquired defects in T cell function. However, it has been a challenge to understand how this infection generates T cell responses and how they mediate parasite control. Here, Cryptosporidium was engineered to express a parasite effector protein (MEDLE-2) that contains the MHC-I restricted SIINFEKL epitope which is recognized by TCR transgenic OT-I CD8 + T cells. These modified parasites induced expansion of endogenous SIINFEKL-specific and OT-I CD8 + T cells that were a source of IFN-γ that could restrict growth of Cryptosporidium . This T cell response was dependent on the translocation of the effector and similar results were observed with another secreted parasite effector (ROP1). Although infection and these translocated effector proteins are restricted to intestinal epithelial cells (IEC), type I dendritic cells (cDC1) were required to generate CD8 + T cell responses to these model antigens. These data sets highlight Cryptosporidium effectors as targets of the immune system and suggest that crosstalk between enterocytes and cDC1s is crucial for CD8 + T cell responses to Cryptosporidium .
Collapse
|
12
|
O'Connor JH, McNamara HA, Cai Y, Coupland LA, Gardiner EE, Parish CR, McMorran BJ, Ganusov VV, Cockburn IA. Interactions with Asialo-Glycoprotein Receptors and Platelets Are Dispensable for CD8 + T Cell Localization in the Murine Liver. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2738-2748. [PMID: 35649630 PMCID: PMC9308657 DOI: 10.4049/jimmunol.2101037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 04/11/2022] [Indexed: 06/15/2023]
Abstract
Liver-resident CD8+ T cells can play critical roles in the control of pathogens, including Plasmodium and hepatitis B virus. Paradoxically, it has also been proposed that the liver may act as the main place for the elimination of CD8+ T cells at the resolution of immune responses. We hypothesized that different adhesion processes may drive residence versus elimination of T cells in the liver. Specifically, we investigated whether the expression of asialo-glycoproteins (ASGPs) drives the localization and elimination of effector CD8+ T cells in the liver, while interactions with platelets facilitate liver residence and protective function. Using murine CD8+ T cells activated in vitro, or in vivo by immunization with Plasmodium berghei sporozoites, we found that, unexpectedly, inhibition of ASGP receptors did not inhibit the accumulation of effector cells in the liver, but instead prevented these cells from accumulating in the spleen. In addition, enforced expression of ASGP on effector CD8+ T cells using St3GalI-deficient cells lead to their loss from the spleen. We also found, using different mouse models of thrombocytopenia, that severe reduction in platelet concentration in circulation did not strongly influence the residence and protective function of CD8+ T cells in the liver. These data suggest that platelets play a marginal role in CD8+ T cell function in the liver. Furthermore, ASGP-expressing effector CD8+ T cells accumulate in the spleen, not the liver, prior to their destruction.
Collapse
Affiliation(s)
- James H O'Connor
- Division of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
- Australian National University Medical School, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Hayley A McNamara
- Division of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Yeping Cai
- Division of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Lucy A Coupland
- Division of Genome Science and Cancer, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia; and
| | - Elizabeth E Gardiner
- Division of Genome Science and Cancer, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia; and
| | - Christopher R Parish
- Division of Genome Science and Cancer, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia; and
| | - Brendan J McMorran
- Division of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Vitaly V Ganusov
- Department of Microbiology, University of Tennessee, Knoxville, TN
| | - Ian A Cockburn
- Division of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia;
| |
Collapse
|
13
|
Arias CF, Acosta FJ, Fernandez-Arias C. Killing the competition: a theoretical framework for liver-stage malaria. Open Biol 2022; 12:210341. [PMID: 35350863 PMCID: PMC8965401 DOI: 10.1098/rsob.210341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The first stage of malaria infections takes place inside the host's hepatocytes. Remarkably, Plasmodium parasites do not infect hepatocytes immediately after reaching the liver. Instead, they migrate through several hepatocytes before infecting their definitive host cells, thus increasing their chances of immune destruction. Considering that malaria can proceed normally without cell traversal, this is indeed a puzzling behaviour. In fact, the role of hepatocyte traversal remains unknown to date, implying that the current understanding of malaria is incomplete. In this work, we hypothesize that the parasites traverse hepatocytes to actively trigger an immune response in the host. This behaviour would be part of a strategy of superinfection exclusion aimed to reduce intraspecific competition during the blood stage of the infection. Based on this hypothesis, we formulate a comprehensive theory of liver-stage malaria that integrates all the available knowledge about the infection. The interest of this new paradigm is not merely theoretical. It highlights major issues in the current empirical approach to the study of Plasmodium and suggests new strategies to fight malaria.
Collapse
Affiliation(s)
- Clemente F. Arias
- Centro de Investigaciones Biológicas (CSIC), Madrid, Spain,Grupo Interdisciplinar de Sistemas Complejos de Madrid, Spain
| | | | - Cristina Fernandez-Arias
- Departamento de Inmunología, Universidad Complutense de Madrid, Spain,Instituto de Medicina Molecular, Universidade de Lisboa, Portugal
| |
Collapse
|
14
|
Kreutzfeld O, Grützke J, Ingmundson A, Müller K, Matuschewski K. Absence of PEXEL-Dependent Protein Export in Plasmodium Liver Stages Cannot Be Restored by Gain of the HSP101 Protein Translocon ATPase. Front Genet 2021; 12:742153. [PMID: 34956312 PMCID: PMC8693896 DOI: 10.3389/fgene.2021.742153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
Host cell remodeling is critical for successful Plasmodium replication inside erythrocytes and achieved by targeted export of parasite-encoded proteins. In contrast, during liver infection the malarial parasite appears to avoid protein export, perhaps to limit exposure of parasite antigens by infected liver cells. HSP101, the force-generating ATPase of the protein translocon of exported proteins (PTEX) is the only component that is switched off during early liver infection. Here, we generated transgenic Plasmodium berghei parasite lines that restore liver stage expression of HSP101. HSP101 expression in infected hepatocytes was achieved by swapping the endogenous promoter with the ptex150 promoter and by inserting an additional copy under the control of the elongation one alpha (ef1α) promoter. Both promoters drive constitutive and, hence, also pre-erythrocytic expression. Transgenic parasites were able to complete the life cycle, but failed to export PEXEL-proteins in early liver stages. Our results suggest that PTEX-dependent early liver stage export cannot be restored by addition of HSP101, indicative of alternative export complexes or other functions of the PTEX core complex during liver infection.
Collapse
Affiliation(s)
- Oriana Kreutzfeld
- Molecular Parasitology, Institute of Biology/Faculty for Life Sciences, Humboldt Universität zu Berlin, Berlin, Germany.,Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany.,Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Josephine Grützke
- Molecular Parasitology, Institute of Biology/Faculty for Life Sciences, Humboldt Universität zu Berlin, Berlin, Germany.,Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany.,Department of Biological Safety, Federal Institute for Risk Assessment, Berlin, Germany
| | - Alyssa Ingmundson
- Molecular Parasitology, Institute of Biology/Faculty for Life Sciences, Humboldt Universität zu Berlin, Berlin, Germany.,Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Katja Müller
- Molecular Parasitology, Institute of Biology/Faculty for Life Sciences, Humboldt Universität zu Berlin, Berlin, Germany.,Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Kai Matuschewski
- Molecular Parasitology, Institute of Biology/Faculty for Life Sciences, Humboldt Universität zu Berlin, Berlin, Germany.,Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
15
|
Flores-Garcia Y, Wang LT, Park M, Asady B, Idris AH, Kisalu NK, Muñoz C, Pereira LS, Francica JR, Seder RA, Zavala F. The P. falciparum CSP repeat region contains three distinct epitopes required for protection by antibodies in vivo. PLoS Pathog 2021; 17:e1010042. [PMID: 34748617 PMCID: PMC8601602 DOI: 10.1371/journal.ppat.1010042] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/18/2021] [Accepted: 10/18/2021] [Indexed: 11/18/2022] Open
Abstract
Rare and potent monoclonal antibodies (mAbs) against the Plasmodium falciparum (Pf) circumsporozoite protein (CSP) on infective sporozoites (SPZ) preferentially bind the PfCSP junctional tetrapeptide NPDP or NVDP minor repeats while cross-reacting with NANP central repeats in vitro. The extent to which each of these epitopes is required for protection in vivo is unknown. Here, we assessed whether junction-, minor repeat- and central repeat-preferring human mAbs (CIS43, L9 and 317 respectively) bound and protected against in vivo challenge with transgenic P. berghei (Pb) SPZ expressing either PfCSP with the junction and minor repeats knocked out (KO), or PbCSP with the junction and minor repeats knocked in (KI). In vivo protection studies showed that the junction and minor repeats are necessary and sufficient for CIS43 and L9 to neutralize KO and KI SPZ, respectively. In contrast, 317 required major repeats for in vivo protection. These data establish that human mAbs can prevent malaria infection by targeting three different protective epitopes (NPDP, NVDP, NANP) in the PfCSP repeat region. This report will inform vaccine development and the use of mAbs to passively prevent malaria. Human monoclonal antibodies are a promising approach for preventing malaria. Highly potent human antibodies show preferential binding to the junction or minor repeat regions of the circumsporozoite protein (CSP) of P. falciparum and cross-react to repetitive (NANP) repeats. The requirement for these binding sites for mediating protection in vivo remains unknown. Here, using transgenic P. berghei parasites expressing PfCSP containing deletions of these junctional or minor epitopes, or PbCSP containing additions of these PfCSP epitopes, we demonstrate these epitopes are necessary and sufficient to mediate protection and don’t require cross-reactivity to the NANP repeats. Our findings establish a mechanism of antibody mediated protection in vivo to prevent malaria and provide the rationale for multi-epitope vaccines to increase the breadth of the antibody responses.
Collapse
Affiliation(s)
- Yevel Flores-Garcia
- Johns Hopkins Bloomberg School of Public Health, Department of Molecular Microbiology and Immunology, Malaria Research Institute, Baltimore, Maryland, United States of America
| | - Lawrence T. Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Minah Park
- Johns Hopkins Bloomberg School of Public Health, Department of Molecular Microbiology and Immunology, Malaria Research Institute, Baltimore, Maryland, United States of America
| | - Beejan Asady
- Johns Hopkins Bloomberg School of Public Health, Department of Molecular Microbiology and Immunology, Malaria Research Institute, Baltimore, Maryland, United States of America
| | - Azza H. Idris
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Neville K. Kisalu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Christian Muñoz
- Medical Technology Department, Faculty of Health Science, University of Antofagasta, Antofagasta, Chile
| | - Lais S. Pereira
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Joseph R. Francica
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Robert A. Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Fidel Zavala
- Johns Hopkins Bloomberg School of Public Health, Department of Molecular Microbiology and Immunology, Malaria Research Institute, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
16
|
Müller K, Gibbins MP, Roberts M, Reyes‐Sandoval A, Hill AVS, Draper SJ, Matuschewski K, Silvie O, Hafalla JCR. Low immunogenicity of malaria pre-erythrocytic stages can be overcome by vaccination. EMBO Mol Med 2021; 13:e13390. [PMID: 33709544 PMCID: PMC8033512 DOI: 10.15252/emmm.202013390] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 11/09/2022] Open
Abstract
Immunogenicity is considered one important criterion for progression of candidate vaccines to further clinical evaluation. We tested this assumption in an infection and vaccination model for malaria pre-erythrocytic stages. We engineered Plasmodium berghei parasites that harbour a well-characterised epitope for stimulation of CD8+ T cells, either as an antigen in the sporozoite surface-expressed circumsporozoite protein or the parasitophorous vacuole membrane associated protein upregulated in sporozoites 4 (UIS4) expressed in exo-erythrocytic forms (EEFs). We show that the antigen origin results in profound differences in immunogenicity with a sporozoite antigen eliciting robust, superior antigen-specific CD8+ T-cell responses, whilst an EEF antigen evokes poor responses. Despite their contrasting immunogenic properties, both sporozoite and EEF antigens gain access to antigen presentation pathways in hepatocytes, as recognition and targeting by vaccine-induced effector CD8+ T cells results in high levels of protection when targeting either antigen. Our study is the first demonstration that poorly immunogenic EEF antigens do not preclude their susceptibility to antigen-specific CD8+ T-cell killing, which has wide-ranging implications on antigen prioritisation for next-generation pre-erythrocytic malaria vaccines.
Collapse
Affiliation(s)
- Katja Müller
- Parasitology UnitMax Planck Institute for Infection BiologyBerlinGermany
- Department of Molecular ParasitologyInstitute of BiologyHumboldt UniversityBerlinGermany
| | - Matthew P Gibbins
- Department of Infection BiologyFaculty of Infectious and Tropical DiseasesLondon School of Hygiene and Tropical MedicineLondonUK
- Present address:
Wellcome Centre for Integrative ParasitologyInstitute of Infection, Immunity and InflammationUniversity of GlasgowGlasgowUK
| | - Mark Roberts
- Department of Infection BiologyFaculty of Infectious and Tropical DiseasesLondon School of Hygiene and Tropical MedicineLondonUK
| | - Arturo Reyes‐Sandoval
- Jenner InstituteUniversity of OxfordOxfordUK
- Present address:
Instituto Politécnico NacionalIPN. Av. Luis Enrique Erro s/n, Unidad Adolfo López MateosMexico CityMexico
| | | | | | - Kai Matuschewski
- Parasitology UnitMax Planck Institute for Infection BiologyBerlinGermany
- Department of Molecular ParasitologyInstitute of BiologyHumboldt UniversityBerlinGermany
| | - Olivier Silvie
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies InfectieusesCIMI‐ParisParisFrance
| | - Julius Clemence R Hafalla
- Department of Infection BiologyFaculty of Infectious and Tropical DiseasesLondon School of Hygiene and Tropical MedicineLondonUK
| |
Collapse
|
17
|
Abstract
Obligate intracellular malaria parasites reside within a vacuolar compartment generated during invasion which is the principal interface between pathogen and host. To subvert their host cell and support their metabolism, these parasites coordinate a range of transport activities at this membrane interface that are critically important to parasite survival and virulence, including nutrient import, waste efflux, effector protein export, and uptake of host cell cytosol. Here, we review our current understanding of the transport mechanisms acting at the malaria parasite vacuole during the blood and liver-stages of development with a particular focus on recent advances in our understanding of effector protein translocation into the host cell by the Plasmodium Translocon of EXported proteins (PTEX) and small molecule transport by the PTEX membrane-spanning pore EXP2. Comparison to Toxoplasma gondii and other related apicomplexans is provided to highlight how similar and divergent mechanisms are employed to fulfill analogous transport activities.
Collapse
Affiliation(s)
- Josh R. Beck
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, United States of America
| | - Chi-Min Ho
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| |
Collapse
|
18
|
Fernandez-Ruiz D, de Menezes MN, Holz LE, Ghilas S, Heath WR, Beattie L. Harnessing liver-resident memory T cells for protection against malaria. Expert Rev Vaccines 2021; 20:127-141. [PMID: 33501877 DOI: 10.1080/14760584.2021.1881485] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Tissue-resident memory T cells (TRM cells) are powerful mediators of protracted adaptive immunity to infection in peripheral organs. Harnessing TRM cells through vaccination hence promises unprecedented potential for protection against infection. A paramount example of this is malaria, a major infectious disease for which immunity through traditional vaccination strategies remains challenging. Liver TRM cells appear to be highly protective against malaria, and recent developments in our knowledge of the biology of these cells have defined promising, novel strategies for their induction. AREAS COVERED Here, we describe the path that led to the discovery of TRM cells and discuss the importance of liver TRM cells in immunity against Plasmodium spp. infection; we summarize current knowledge on TRM cell biology and discuss the current state and potential of TRM-based vaccination against malaria. EXPERT OPINION TRM based vaccination has emerged as a promising means to achieve efficient protection against malaria. Recent advances provide a solid basis for continuing the development of this area of research. Deeper understanding of the mechanisms that mediate TRM formation and maintenance and identification of immunogenic and protective target epitopes suitable for human vaccination remain the main challenges for translation of these discoveries.
Collapse
Affiliation(s)
- Daniel Fernandez-Ruiz
- Dept. Of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Vic, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne,Vic, Australia
| | - Maria N de Menezes
- Dept. Of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Vic, Australia
| | - Lauren E Holz
- Dept. Of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Vic, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne,Vic, Australia
| | - Sonia Ghilas
- Dept. Of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Vic, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne,Vic, Australia
| | - William R Heath
- Dept. Of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Vic, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne,Vic, Australia
| | - Lynette Beattie
- Dept. Of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Vic, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne,Vic, Australia
| |
Collapse
|
19
|
Chatterjee D, Cockburn IA. The challenges of a circumsporozoite protein-based malaria vaccine. Expert Rev Vaccines 2021; 20:113-125. [PMID: 33554669 DOI: 10.1080/14760584.2021.1874924] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION A safe and effective vaccine will likely be necessary for the control or eradication of malaria which kills 400,000 annually. Our most advanced vaccine candidate to date is RTS,S which is based on the Plasmodium falciparum circumsporozoite protein (PfCSP) of the malaria parasite. However, protection by RTS,S is incomplete and short-lived. AREAS COVERED Here we summarize results from recent clinical trials of RTS,S and critically evaluate recent studies that aim to understand the correlates of protective immunity and why vaccine-induced protection is short-lived. In particular, recent systems serology studies have highlighted a key role for the necessity of inducing functional antibodies. In-depth analyses of immune responses to CSP in both mouse models and vaccinated humans have also highlighted difficulties in generating the maintaining high-quality antibody responses. Finally, in recent years biophysical and structural studies of antibody binding to PfCSP have led to a better understanding of how highly potent antibodies can block infection, which can inform vaccine design. EXPERT OPINION We highlight how both structure-guided vaccine design and a better understanding of the immune response to PfCSP can inform a second generation of PfCSP-based vaccines stimulating a broader range of protective targets within PfCSP.
Collapse
Affiliation(s)
- Deepyan Chatterjee
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, the Australian National University, Canberra, Australia
| | - Ian Andrew Cockburn
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, the Australian National University, Canberra, Australia
| |
Collapse
|
20
|
Abuga KM, Jones-Warner W, Hafalla JCR. Immune responses to malaria pre-erythrocytic stages: Implications for vaccine development. Parasite Immunol 2020; 43:e12795. [PMID: 32981095 PMCID: PMC7612353 DOI: 10.1111/pim.12795] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 08/26/2020] [Accepted: 09/14/2020] [Indexed: 12/16/2022]
Abstract
Radiation-attenuated sporozoites induce sterilizing immunity and remain the 'gold standard' for malaria vaccine development. Despite practical challenges in translating these whole sporozoite vaccines to large-scale intervention programmes, they have provided an excellent platform to dissect the immune responses to malaria pre-erythrocytic (PE) stages, comprising both sporozoites and exoerythrocytic forms. Investigations in rodent models have provided insights that led to the clinical translation of various vaccine candidates-including RTS,S/AS01, the most advanced candidate currently in a trial implementation programme in three African countries. With advances in immunology, transcriptomics and proteomics, and application of lessons from past failures, an effective, long-lasting and wide-scale malaria PE vaccine remains feasible. This review underscores the progress in PE vaccine development, focusing on our understanding of host-parasite immunological crosstalk in the tissue environments of the skin and the liver. We highlight possible gaps in the current knowledge of PE immunity that can impact future malaria vaccine development efforts.
Collapse
Affiliation(s)
- Kelvin Mokaya Abuga
- Department of Infection Biology, Faculty of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, UK.,Department of Epidemiology and Demography, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - William Jones-Warner
- Department of Infection Biology, Faculty of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Julius Clemence R Hafalla
- Department of Infection Biology, Faculty of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
21
|
Importance of the Immunodominant CD8 + T Cell Epitope of Plasmodium berghei Circumsporozoite Protein in Parasite- and Vaccine-Induced Protection. Infect Immun 2020; 88:IAI.00383-20. [PMID: 32719159 DOI: 10.1128/iai.00383-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 07/22/2020] [Indexed: 12/15/2022] Open
Abstract
The circumsporozoite protein (CSP) builds up the surface coat of sporozoites and is the leading malaria pre-erythrocytic-stage vaccine candidate. CSP has been shown to induce robust CD8+ T cell responses that are capable of eliminating developing parasites in hepatocytes, resulting in protective immunity. In this study, we characterized the importance of the immunodominant CSP-derived epitope SYIPSAEKI of Plasmodium berghei in both sporozoite- and vaccine-induced protection in murine infection models. In BALB/c mice, where SYIPSAEKI is efficiently presented in the context of the major histocompatibility complex class I (MHC-I) molecule H-2-Kd, we established that epitope-specific CD8+ T cell responses contribute to parasite killing following sporozoite immunization. Yet, sterile protection was achieved in the absence of this epitope, substantiating the concept that other antigens can be sufficient for parasite-induced protective immunity. Furthermore, we demonstrated that SYIPSAEKI-specific CD8+ T cell responses elicited by viral-vectored CSP-expressing vaccines effectively targeted parasites in hepatocytes. The resulting sterile protection strictly relied on the expression of SYIPSAEKI. In C57BL/6 mice, which are unable to present the immunodominant epitope, CSP-based vaccines did not confer complete protection, despite the induction of high levels of CSP-specific antibodies. These findings underscore the significance of CSP in protection against malaria pre-erythrocytic stages and demonstrate that a significant proportion of the protection against the parasite is mediated by CD8+ T cells specific for the immunodominant CSP-derived epitope.
Collapse
|
22
|
Heide J, Vaughan KC, Sette A, Jacobs T, Schulze Zur Wiesch J. Comprehensive Review of Human Plasmodium falciparum-Specific CD8+ T Cell Epitopes. Front Immunol 2019; 10:397. [PMID: 30949162 PMCID: PMC6438266 DOI: 10.3389/fimmu.2019.00397] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/15/2019] [Indexed: 12/16/2022] Open
Abstract
Control of malaria is an important global health issue and there is still an urgent need for the development of an effective prophylactic vaccine. Multiple studies have provided strong evidence that Plasmodium falciparum-specific MHC class I-restricted CD8+ T cells are important for sterile protection against Plasmodium falciparum infection. Here, we present an interactive epitope map of all P. falciparum-specific CD8+ T cell epitopes published to date, based on a comprehensive data base (IEDB), and literature search. The majority of the described P. falciparum-specific CD8+ T cells were directed against the antigens CSP, TRAP, AMA1, and LSA1. Notably, most of the epitopes were discovered in vaccine trials conducted with malaria-naïve volunteers. Only few immunological studies of P. falciparum-specific CD8+ T cell epitopes detected in patients suffering from acute malaria or in people living in malaria endemic areas have been published. Further detailed immunological mappings of P. falciparum-specific epitopes of a broader range of P. falciparum proteins in different settings and with different disease status are needed to gain a more comprehensive understanding of the role of CD8+ T cell responses for protection, and to better guide vaccine design and to study their efficacy.
Collapse
Affiliation(s)
- Janna Heide
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Kerrie C Vaughan
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States.,Department of Medicine, Division of Infectious Diseases, University of California, San Diego (UCSD), La Jolla, CA, United States
| | - Thomas Jacobs
- Protozoa Immunology, Bernhard-Nocht-Institute of Tropical Medicine, Hamburg, Germany
| | - Julian Schulze Zur Wiesch
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| |
Collapse
|
23
|
Walk J, Stok JE, Sauerwein RW. Can Patrolling Liver-Resident T Cells Control Human Malaria Parasite Development? Trends Immunol 2019; 40:186-196. [PMID: 30713008 DOI: 10.1016/j.it.2019.01.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/08/2019] [Accepted: 01/13/2019] [Indexed: 12/23/2022]
Abstract
Recently, a population of non-recirculating, tissue-resident memory CD8+ T cells has been identified; cells that seems to act as key sentinels for invading microorganisms with enhanced effector functions. In malaria, the liver represents the first site for parasite development before a definite infection is established in circulating red blood cells. Here, we discuss the evidence obtained from animal models on several diseases and hypothesize that liver-resident memory CD8+ T cells (hepatic TRM) play a critical role in providing protective liver-stage immunity against Plasmodium malaria parasites. Although observations in human malaria trials are limited to peripheral blood, we propose recommendations for the translation of some of these findings to human malaria research.
Collapse
Affiliation(s)
- Jona Walk
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands; Radboud Center for Infectious Diseases, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Jorn E Stok
- University Medical Center Utrecht, PO Box 85500, Utrecht, The Netherlands
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands; Radboud Center for Infectious Diseases, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
24
|
Cockburn IA, Seder RA. Malaria prevention: from immunological concepts to effective vaccines and protective antibodies. Nat Immunol 2018; 19:1199-1211. [PMID: 30333613 DOI: 10.1038/s41590-018-0228-6] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/31/2018] [Indexed: 02/08/2023]
Abstract
Development of a malaria vaccine remains a critical priority to decrease clinical disease and mortality and facilitate eradication. Accordingly, RTS,S, a protein-subunit vaccine, has completed phase III clinical trials and confers ~30% protection against clinical infection over 4 years. Whole-attenuated-sporozoite and viral-subunit vaccines induce between 20% and 100% protection against controlled human malaria infection, but there is limited published evidence to date for durable, high-level efficacy (>50%) against natural exposure. Importantly, fundamental scientific advances related to the potency, durability, breadth and location of immune responses will be required for improving vaccine efficacy with these and other vaccine approaches. In this Review, we focus on the current understanding of immunological mechanisms of protection from animal models and human vaccine studies, and on how these data should inform the development of next-generation vaccines. Furthermore, we introduce the concept of using passive immunization with monoclonal antibodies as a new approach to prevent and eliminate malaria.
Collapse
Affiliation(s)
- Ian A Cockburn
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
25
|
Frank R, Gabel M, Heiss K, Mueller AK, Graw F. Varying Immunizations With Plasmodium Radiation-Attenuated Sporozoites Alter Tissue-Specific CD8 + T Cell Dynamics. Front Immunol 2018; 9:1137. [PMID: 29892289 PMCID: PMC5985394 DOI: 10.3389/fimmu.2018.01137] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 05/07/2018] [Indexed: 12/12/2022] Open
Abstract
Whole sporozoite vaccines represent one of the most promising strategies to induce protection against malaria. However, the development of efficient vaccination protocols still remains a major challenge. To understand how the generation of immunity is affected by variations in vaccination dosage and frequency, we systematically analyzed intrasplenic and intrahepatic CD8+ T cell responses following varied immunizations of mice with radiation-attenuated sporozoites. By combining experimental data and mathematical modeling, our analysis indicates a reversing role of spleen and liver in the generation of protective liver-resident CD8+ T cells during priming and booster injections: While the spleen acts as a critical source compartment during priming, the increase in vaccine-induced hepatic T cell levels is likely due to local reactivation in the liver in response to subsequent booster injections. Higher dosing accelerates the efficient generation of liver-resident CD8+ T cells by especially affecting their local reactivation. In addition, we determine the differentiation and migration pathway from splenic precursors toward hepatic memory cells thereby presenting a mechanistic framework for the impact of various vaccination protocols on these dynamics. Thus, our work provides important insights into organ-specific CD8+ T cell dynamics and their role and interplay in the formation of protective immunity against malaria.
Collapse
Affiliation(s)
- Roland Frank
- Centre for Infectious Diseases, Parasitology Unit, University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Gabel
- Centre for Modeling and Simulation in the Biosciences, BioQuant-Center, Heidelberg University, Heidelberg, Germany
| | - Kirsten Heiss
- Centre for Infectious Diseases, Parasitology Unit, University Hospital Heidelberg, Heidelberg, Germany
| | - Ann-Kristin Mueller
- Centre for Infectious Diseases, Parasitology Unit, University Hospital Heidelberg, Heidelberg, Germany.,German Center for Infection Research (DZIF), Heidelberg, Germany
| | - Frederik Graw
- Centre for Modeling and Simulation in the Biosciences, BioQuant-Center, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
26
|
Nonspecific CD8 + T Cells and Dendritic Cells/Macrophages Participate in Formation of CD8 + T Cell-Mediated Clusters against Malaria Liver-Stage Infection. Infect Immun 2018; 86:IAI.00717-17. [PMID: 29426043 DOI: 10.1128/iai.00717-17] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/25/2018] [Indexed: 12/12/2022] Open
Abstract
CD8+ T cells are the major effector cells that protect against malaria liver-stage infection, forming clusters around Plasmodium-infected hepatocytes and eliminating parasites after a prolonged interaction with these hepatocytes. We aimed to investigate the roles of specific and nonspecific CD8+ T cells in cluster formation and protective immunity. To this end, we used Plasmodium berghei ANKA expressing ovalbumin as well as CD8+ T cells from transgenic mice expressing a T cell receptor specific for ovalbumin (OT-I) and CD8+ T cells specific for an unrelated antigen, respectively. While antigen-specific CD8+ T cells were essential for cluster formation, both antigen-specific and nonspecific CD8+ T cells joined the clusters. However, nonspecific CD8+ T cells did not significantly contribute to protective immunity. In the livers of infected mice, specific CD8+ T cells expressed high levels of CD25, compatible with a local, activated effector phenotype. In vivo imaging of the liver revealed that specific CD8+ T cells interact with CD11c+ cells around infected hepatocytes. The depletion of CD11c+ cells virtually eliminated the clusters in the liver, leading to a significant decrease in protection. These experiments reveal an essential role of hepatic CD11c+ dendritic cells and presumably macrophages in the formation of CD8+ T cell clusters around Plasmodium-infected hepatocytes. Once cluster formation is triggered by parasite-specific CD8+ T cells, specific and unrelated activated CD8+ T cells join the clusters in a chemokine- and dendritic cell-dependent manner. Nonspecific CD8+ T cells seem to play a limited role in protective immunity against Plasmodium parasites.
Collapse
|
27
|
Parmar R, Patel H, Yadav N, Parikh R, Patel K, Mohankrishnan A, Bhurani V, Joshi U, Dalai SK. Infectious Sporozoites of Plasmodium berghei Effectively Activate Liver CD8α + Dendritic Cells. Front Immunol 2018; 9:192. [PMID: 29472929 PMCID: PMC5809440 DOI: 10.3389/fimmu.2018.00192] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 01/23/2018] [Indexed: 11/13/2022] Open
Abstract
Immunization with radiation-attenuated sporozoites (RAS) shown to confer complete sterile protection against Plasmodia liver-stage (LS) infection that lasts about 6 to 9 months in mice. We have found that the intermittent infectious sporozoite challenge to immune mice following RAS vaccination extends the longevity of sterile protection by maintaining CD8+ T cell memory responses to LS infection. It is reported that CD8α+ dendritic cells (DCs) are involved in the induction of LS-specific CD8+ T cells following RAS or genetically attenuated parasite (GAP) vaccination. In this study, we demonstrate that CD8α+ DCs respond differently to infectious sporozoite or RAS inoculation. The higher accumulation and activation of CD8α+ DCs was seen in the liver in response to infectious sporozoite 72 h postinoculation and found to be associated with higher expression of chemokines (CCL-20 and CCL-21) and type I interferon response via toll-like receptor signaling in liver. Moreover, the infectious sporozoites were found to induce qualitative changes in terms of the increased MHCII expression as well as costimulatory molecules including CD40 on the CD8α+ DCs compared to RAS inoculation. We have also found that infectious sporozoite challenge increased CD40L-expressing CD4+ T cells, which could help CD8+ T cells in the liver through "licensing" of the antigen-presenting cells. Our results suggest that infectious sporozoite challenge to prior RAS immunized mice modulates the CD8α+ DCs, which might be shaping the fate of memory CD8+ T cells against Plasmodium LS infection.
Collapse
Affiliation(s)
- Rajesh Parmar
- Institute of Science, Nirma University, Ahmedabad, India
| | - Hardik Patel
- Institute of Science, Nirma University, Ahmedabad, India
| | - Naveen Yadav
- Institute of Science, Nirma University, Ahmedabad, India
| | - Ritika Parikh
- Institute of Science, Nirma University, Ahmedabad, India
| | - Khyati Patel
- Institute of Science, Nirma University, Ahmedabad, India
| | | | | | - Urja Joshi
- Institute of Science, Nirma University, Ahmedabad, India
| | | |
Collapse
|
28
|
Silvie O, Amino R, Hafalla JC. Tissue-specific cellular immune responses to malaria pre-erythrocytic stages. Curr Opin Microbiol 2017; 40:160-167. [PMID: 29217460 DOI: 10.1016/j.mib.2017.12.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/30/2017] [Accepted: 12/01/2017] [Indexed: 11/30/2022]
Abstract
Complete and long-lasting protective immunity against malaria can be achieved through vaccination with invasive live attenuated Plasmodium sporozoites, the motile stage inoculated in the host skin during a mosquito bite. Protective immunity relies primarily on effector CD8+ T cells targeting the parasite in the liver. Understanding the tissue-specific features of the immune response is emerging as a vital requirement for understanding protective immunity. The small parasite inoculum, the scarcity of infected cells and the tolerogenic properties of the liver represent hurdles for the establishment of protective immunity in endemic areas. In this review, we discuss recent advances on liver-specific features of immunity including innate recognition of malaria pre-erythrocytic stages, CD8+ T cell interactions with infected hepatocytes, antigen presentation for effective CD8+ T cell responses and generation of liver-resident memory CD8+ T cells. A better understanding of the factors involved in the induction and maintenance of effector CD8+ T cell immunity against malaria pre-erythrocytic stages is crucial for the development of an effective vaccine targeting the initial phase of malaria infection.
Collapse
Affiliation(s)
- Olivier Silvie
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, U1135, ERL8255, Paris, France.
| | - Rogerio Amino
- Unit of Malaria Infection and Immunity, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France.
| | - Julius Clemence Hafalla
- Immunology and Infection Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom.
| |
Collapse
|
29
|
Baleeiro RB, Walden P. Immature human DCs efficiently translocate endocytosed antigens into the cytosol for proteasomal processing. Mol Immunol 2017. [PMID: 28644974 DOI: 10.1016/j.molimm.2017.06.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Cross-presentation of endocytosed antigen is essential for induction of CD8 effector T cell responses and a hallmark of dendritic cells (DCs). The mode of antigen processing in this context is controversial and some models imply translocation of the antigen from the endosomes into the cytosol. To test this hypothesis we made use of the pro-apoptotic properties of cytochrome c when in the cytosol, and confirmed that it indeed triggered apoptosis of human immature DCs but only at high concentrations. Proteasome inhibitors reduced the required concentration of cytochrome c thousand-fold, indicating that protein translocated into the cytosol is rapidly degraded by proteasomes. Mature DCs were also susceptible to cytochrome c-triggered apoptosis at high concentrations but proteasome inhibitors did not increase their sensitivity. Other cross-presenting cells such as B cells and monocytes were not sensitive to cytochrome c at all, indicating that they do not shuttle internalized antigen into the cytosol. Thus, processing of internalized antigens seems to follow different pathways depending on cell type and, in case of DCs, maturation state. Immature DCs appear to have a unique capacity to shuttle external antigen into the cytosol for proteasomal processing, which could explain their efficiency in antigen cross-presentation.
Collapse
Affiliation(s)
- Renato B Baleeiro
- Charité-Universitätsmedizin Berlin, Department of Dermatology, Venerology and Allergology, Clinical Research Group Tumour Immunology, Berlin, Germany
| | - Peter Walden
- Charité-Universitätsmedizin Berlin, Department of Dermatology, Venerology and Allergology, Clinical Research Group Tumour Immunology, Berlin, Germany.
| |
Collapse
|
30
|
Kreutzfeld O, Müller K, Matuschewski K. Engineering of Genetically Arrested Parasites (GAPs) For a Precision Malaria Vaccine. Front Cell Infect Microbiol 2017; 7:198. [PMID: 28620583 PMCID: PMC5450620 DOI: 10.3389/fcimb.2017.00198] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 05/04/2017] [Indexed: 01/08/2023] Open
Abstract
Continuous stage conversion and swift changes in the antigenic repertoire in response to acquired immunity are hallmarks of complex eukaryotic pathogens, including Plasmodium species, the causative agents of malaria. Efficient elimination of Plasmodium liver stages prior to blood infection is one of the most promising malaria vaccine strategies. Here, we describe different genetically arrested parasites (GAPs) that have been engineered in Plasmodium berghei, P. yoelii and P. falciparum and compare their vaccine potential. A better understanding of the immunological mechanisms of prime and boost by arrested sporozoites and experimental strategies to enhance vaccine efficacy by further engineering existing GAPs into a more immunogenic form hold promise for continuous improvements of GAP-based vaccines. A critical hurdle for vaccines that elicit long-lasting protection against malaria, such as GAPs, is safety and efficacy in vulnerable populations. Vaccine research should focus on solutions toward turning malaria into a vaccine-preventable disease, which would offer an exciting new path of malaria control.
Collapse
Affiliation(s)
- Oriana Kreutzfeld
- Department of Molecular Parasitology, Institute of Biology, Humboldt UniversityBerlin, Germany
| | - Katja Müller
- Department of Molecular Parasitology, Institute of Biology, Humboldt UniversityBerlin, Germany
| | - Kai Matuschewski
- Department of Molecular Parasitology, Institute of Biology, Humboldt UniversityBerlin, Germany
| |
Collapse
|
31
|
Othman AS, Marin-Mogollon C, Salman AM, Franke-Fayard BM, Janse CJ, Khan SM. The use of transgenic parasites in malaria vaccine research. Expert Rev Vaccines 2017; 16:1-13. [DOI: 10.1080/14760584.2017.1333426] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Ahmad Syibli Othman
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
- Faculty of Health Sciences, Universiti Sultan Zainal Abidin, Terengganu, Malaysia
| | - Catherin Marin-Mogollon
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | | | - Blandine M. Franke-Fayard
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Chris J. Janse
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Shahid M. Khan
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| |
Collapse
|
32
|
Spencer AJ, Longley RJ, Gola A, Ulaszewska M, Lambe T, Hill AVS. The Threshold of Protection from Liver-Stage Malaria Relies on a Fine Balance between the Number of Infected Hepatocytes and Effector CD8 + T Cells Present in the Liver. THE JOURNAL OF IMMUNOLOGY 2017; 198:2006-2016. [PMID: 28087668 PMCID: PMC5318841 DOI: 10.4049/jimmunol.1601209] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 12/16/2016] [Indexed: 12/20/2022]
Abstract
Since the demonstration of sterile protection afforded by injection of irradiated sporozoites, CD8+ T cells have been shown to play a significant role in protection from liver-stage malaria. This is, however, dependent on the presence of an extremely high number of circulating effector cells, thought to be necessary to scan, locate, and kill infected hepatocytes in the short time that parasites are present in the liver. We used an adoptive transfer model to elucidate the kinetics of the effector CD8+ T cell response in the liver following Plasmodium berghei sporozoite challenge. Although effector CD8+ T cells require <24 h to find, locate, and kill infected hepatocytes, active migration of Ag-specific CD8+ T cells into the liver was not observed during the 2-d liver stage of infection, as divided cells were only detected from day 3 postchallenge. However, the percentage of donor cells recruited into division was shown to indicate the level of Ag presentation from infected hepatocytes. By titrating the number of transferred Ag-specific effector CD8+ T cells and sporozoites, we demonstrate that achieving protection toward liver-stage malaria is reliant on CD8+ T cells being able to locate infected hepatocytes, resulting in a protection threshold dependent on a fine balance between the number of infected hepatocytes and CD8+ T cells present in the liver. With such a fine balance determining protection, achieving a high number of CD8+ T cells will be critical to the success of a cell-mediated vaccine against liver-stage malaria.
Collapse
Affiliation(s)
| | - Rhea J Longley
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Anita Gola
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Marta Ulaszewska
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Teresa Lambe
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Adrian V S Hill
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, United Kingdom
| |
Collapse
|
33
|
Angiotensin II type-1 receptor (AT 1R) regulates expansion, differentiation, and functional capacity of antigen-specific CD8 + T cells. Sci Rep 2016; 6:35997. [PMID: 27782175 PMCID: PMC5080615 DOI: 10.1038/srep35997] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 10/10/2016] [Indexed: 12/25/2022] Open
Abstract
Angiotensin II (Ang II) and its receptor AT1 (AT1R), an important effector axis of renin-angiotensin system (RAS), have been demonstrated to regulate T-cell responses. However, these studies characterized Ang II and AT1R effects using pharmacological tools, which do not target only Ang II/AT1R axis. The specific role of AT1R expressed by antigen-specific CD8+ T cells is unknown. Then we immunized transgenic mice expressing a T-cell receptor specific for SIINFEKL epitope (OT-I mice) with sporozoites of the rodent malaria parasite Plasmodium berghei expressing the cytotoxic epitope SIINFEKL. Early priming events after immunization were not affected but the expansion and contraction of AT1R-deficient (AT1R-/-) OT-I cells was decreased. Moreover, they seemed more activated, express higher levels of CTLA-4, PD-1, LAG-3, and have decreased functional capacity during the effector phase. Memory AT1R-/- OT-I cells exhibited higher IL-7Rα expression, activation, and exhaustion phenotypes but less cytotoxic capacity. Importantly, AT1R-/- OT-I cells show better control of blood parasitemia burden and ameliorate mice survival during lethal disease induced by blood-stage malaria. Our study reveals that AT1R in antigen-specific CD8+ T cells regulates expansion, differentiation, and function during effector and memory phases of the response against Plasmodium, which could apply to different infectious agents.
Collapse
|
34
|
Singer M, Frischknecht F. Time for Genome Editing: Next-Generation Attenuated Malaria Parasites. Trends Parasitol 2016; 33:202-213. [PMID: 27793562 DOI: 10.1016/j.pt.2016.09.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/20/2016] [Accepted: 09/26/2016] [Indexed: 12/20/2022]
Abstract
Immunization with malaria parasites that developmentally arrest in or immediately after the liver stage is the only way currently known to confer sterilizing immunity in both humans and rodent models. There are various ways to attenuate parasite development resulting in different timings of arrest, which has a significant impact on vaccination efficiency. To understand what most impacts vaccination efficiency, newly developed gain-of-function methods can now be used to generate a wide array of differently attenuated parasites. The combination of multiple attenuation approaches offers the potential to engineer efficiently attenuated Plasmodium parasites and learn about their fascinating biology at the same time. Here we discuss recent studies and the potential of targeted parasite manipulation using genome editing to develop live attenuated malaria vaccines.
Collapse
Affiliation(s)
- Mirko Singer
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany.
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany.
| |
Collapse
|
35
|
Protective immunity to liver-stage malaria. Clin Transl Immunology 2016; 5:e105. [PMID: 27867517 PMCID: PMC5099428 DOI: 10.1038/cti.2016.60] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 09/14/2016] [Accepted: 09/14/2016] [Indexed: 12/15/2022] Open
Abstract
Despite decades of research and recent clinical trials, an efficacious long-lasting preventative vaccine for malaria remains elusive. This parasite infects mammals via mosquito bites, progressing through several stages including the relatively short asymptomatic liver stage followed by the more persistent cyclic blood stage, the latter of which is responsible for all disease symptoms. As the liver acts as a bottleneck to blood-stage infection, it represents a potential site for parasite and disease control. In this review, we discuss immunity to liver-stage malaria. It is hoped that the knowledge gained from animal models of malaria immunity will translate into a more powerful and effective vaccine to reduce this global health problem.
Collapse
|
36
|
Cockburn IA, Zavala F. Dendritic cell function and antigen presentation in malaria. Curr Opin Immunol 2016; 40:1-6. [DOI: 10.1016/j.coi.2016.01.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 01/18/2016] [Indexed: 10/22/2022]
|
37
|
Doll KL, Pewe LL, Kurup SP, Harty JT. Discriminating Protective from Nonprotective Plasmodium-Specific CD8+ T Cell Responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 196:4253-62. [PMID: 27084099 PMCID: PMC4868661 DOI: 10.4049/jimmunol.1600155] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/19/2016] [Indexed: 11/19/2022]
Abstract
Despite decades of research, malaria remains a global health crisis. Current subunit vaccine approaches do not provide efficient long-term, sterilizing immunity against Plasmodium infections in humans. Conversely, whole parasite vaccinations with their larger array of target Ags have conferred long-lasting sterilizing protection to humans. Similar studies in rodent models of malaria reveal that CD8(+) T cells play a critical role in liver-stage immunity after whole parasite vaccination. However, it is unknown whether all CD8(+) T cell specificities elicited by whole parasite vaccination contribute to protection, an issue of great relevance for enhanced subunit vaccination. In this article, we show that robust CD8(+) T cell responses of similar phenotype are mounted after prime-boost immunization against Plasmodium berghei glideosome-associated protein 5041-48-, sporozoite-specific protein 20318-325-, thrombospondin-related adhesion protein (TRAP) 130-138-, or circumsporozoite protein (CSP) 252-260-derived epitopes in mice, but only CSP252-260- and TRAP130-138-specific CD8(+) T cells provide sterilizing immunity and reduce liver parasite burden after sporozoite challenge. Further, CD8(+) T cells specific to sporozoite surface-expressed CSP and TRAP proteins, but not intracellular glideosome-associated protein 50 and sporozoite-specific protein 20, efficiently recognize sporozoite-infected hepatocytes in vitro. These results suggest that: 1) protection-relevant antigenic targets, regardless of their immunogenic potential, must be efficiently presented by infected hepatocytes for CD8(+) T cells to eliminate liver-stage Plasmodium infection; and 2) proteins expressed on the surface of sporozoites may be good target Ags for protective CD8(+) T cells.
Collapse
Affiliation(s)
- Katherine L Doll
- Department of Microbiology, University of Iowa, Iowa City, IA 52242
| | - Lecia L Pewe
- Department of Microbiology, University of Iowa, Iowa City, IA 52242
| | | | - John T Harty
- Department of Microbiology, University of Iowa, Iowa City, IA 52242; Department of Pathology, University of Iowa, Iowa City, IA 52242; and Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
38
|
Pichugin A, Steers N, De La Vega P, Zarling S, Chalom I, Krzych U. TAP-mediated processing of exoerythrocytic antigens is essential for protection induced with radiation-attenuated Plasmodium sporozoites. Eur J Immunol 2016; 46:885-96. [PMID: 26703789 DOI: 10.1002/eji.201545748] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 11/25/2015] [Accepted: 12/18/2015] [Indexed: 02/01/2023]
Abstract
MHC class I dependent CD8(+) T cells are essential for protection induced by radiation-attenuated Plasmodium sporozoites (RAS) in murine malaria models. Apart from the mechanism of activation of CD8(+) T cells specific for the circumsporozoite protein, the major sporozoite antigen (Ag), CD8(+) T cells specific for other exoerythrocytic Ags that have been shown to mediate protection have not been thoroughly investigated. Specifically, mechanisms of processing and presentation of exoerythrocytic Ags, which includes liver stage (LS) Ags, remain poorly understood. We hypothesize that as exogenous proteins, LS Ags are processed by mechanisms involving either the TAP-dependent phagosomal-to-cytosol or TAP-independent vacuolar pathway of cross-presentation. We used TAP-deficient mice to investigate whether LS Ag mediated induction of naïve CD8(+) T cells and their recall during sporozoite challenge occur by the TAP-dependent or TAP-independent pathways. On the basis of functional attributes, CD8(+) T cells were activated via the TAP-independent pathway during immunizations with Plasmodium berghei RAS; however, IFN-γ(+) CD8(+) T cells previously induced by P. berghei RAS in TAP-deficient mice failed to be recalled against sporozoite challenge and the mice became parasitemic. On the basis of these observations, we propose that TAP-associated Ag processing is indispensable for sterile protection induced with P. berghei RAS.
Collapse
Affiliation(s)
- Alexander Pichugin
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Nick Steers
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Patricia De La Vega
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Stasya Zarling
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Isaac Chalom
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Urszula Krzych
- Department of Cellular Immunology, Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| |
Collapse
|
39
|
Powles L, Xiang SD, Selomulya C, Plebanski M. The Use of Synthetic Carriers in Malaria Vaccine Design. Vaccines (Basel) 2015; 3:894-929. [PMID: 26529028 PMCID: PMC4693224 DOI: 10.3390/vaccines3040894] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 09/28/2015] [Accepted: 10/16/2015] [Indexed: 11/29/2022] Open
Abstract
Malaria vaccine research has been ongoing since the 1980s with limited success. However, recent improvements in our understanding of the immune responses required to combat each stage of infection will allow for intelligent design of both antigens and their associated delivery vaccine vehicles/vectors. Synthetic carriers (also known as vectors) are usually particulate and have multiple properties, which can be varied to control how an associated vaccine interacts with the host, and consequently how the immune response develops. This review comprehensively analyzes both historical and recent studies in which synthetic carriers are used to deliver malaria vaccines. Furthermore, the requirements for a synthetic carrier, such as size, charge, and surface chemistry are reviewed in order to understand the design of effective particle-based vaccines against malaria, as well as providing general insights. Synthetic carriers have the ability to alter and direct the immune response, and a better control of particle properties will facilitate improved vaccine design in the near future.
Collapse
Affiliation(s)
- Liam Powles
- Department of Chemical Engineering, Monash University, Clayton, VIC 3800, Australia.
| | - Sue D Xiang
- Department of Immunology and Pathology, Monash University, Melbourne, VIC 3004, Australia.
- Therapeutics and Regenerative Medicine Division, The Monash Institute of Medical Engineering (MIME), Monash University, Clayton, VIC 3800, Australia.
| | - Cordelia Selomulya
- Department of Chemical Engineering, Monash University, Clayton, VIC 3800, Australia.
| | - Magdalena Plebanski
- Department of Immunology and Pathology, Monash University, Melbourne, VIC 3004, Australia.
- Therapeutics and Regenerative Medicine Division, The Monash Institute of Medical Engineering (MIME), Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
40
|
Kalanon M, Bargieri D, Sturm A, Matthews K, Ghosh S, Goodman CD, Thiberge S, Mollard V, McFadden GI, Ménard R, Koning‐Ward TF. The
Plasmodium
translocon of exported proteins component EXP2 is critical for establishing a patent malaria infection in mice. Cell Microbiol 2015; 18:399-412. [DOI: 10.1111/cmi.12520] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 08/26/2015] [Accepted: 08/31/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Ming Kalanon
- Molecular and Medical Research Unit, School of MedicineDeakin University Waurn Ponds Geelong Victoria 3216 Australia
| | - Daniel Bargieri
- Unité de Biologie et Génétique du PaludismeInstitut Pasteur Paris France
- Department of Parasitology, Institute of Biomedical SciencesUniversity of São Paulo São Paulo SP Brazil
| | - Angelika Sturm
- School of BioSciencesThe University of Melbourne Parkville Victoria 3010 Australia
| | - Kathryn Matthews
- Molecular and Medical Research Unit, School of MedicineDeakin University Waurn Ponds Geelong Victoria 3216 Australia
| | - Sreejoyee Ghosh
- Molecular and Medical Research Unit, School of MedicineDeakin University Waurn Ponds Geelong Victoria 3216 Australia
| | | | - Sabine Thiberge
- Unité de Biologie et Génétique du PaludismeInstitut Pasteur Paris France
| | - Vanessa Mollard
- School of BioSciencesThe University of Melbourne Parkville Victoria 3010 Australia
| | - Geoffrey I. McFadden
- School of BioSciencesThe University of Melbourne Parkville Victoria 3010 Australia
| | - Robert Ménard
- Unité de Biologie et Génétique du PaludismeInstitut Pasteur Paris France
| | - Tania F. Koning‐Ward
- Molecular and Medical Research Unit, School of MedicineDeakin University Waurn Ponds Geelong Victoria 3216 Australia
| |
Collapse
|
41
|
Montagna GN, Biswas A, Hildner K, Matuschewski K, Dunay IR. Batf3 deficiency proves the pivotal role of CD8α + dendritic cells in protection induced by vaccination with attenuated Plasmodium sporozoites. Parasite Immunol 2015; 37:533-543. [PMID: 26284735 DOI: 10.1111/pim.12222] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 08/10/2015] [Indexed: 12/28/2022]
Abstract
Increasing evidence indicates that hepatic CD8α+ dendritic cells (DCs) are important antigen cross-presenting cells (APC) involved in the priming of protective CD8+ T-cell responses induced by live-attenuated Plasmodium sporozoites. Experimental proof for a critical role of CD8α+ DCs in protective pre-erythrocytic malaria immunizations has pivotal implications for vaccine development, including improved vectored subunit vaccines. Employing Batf3-/- mice, which lack functional CD8α+ DCs, we demonstrate that deficiency of these particular APCs completely abolishes protection and corresponding signatures of vaccine-induced immunity. We show that in wild-type, but not in Batf3-/- , mice CD8α+ DCs accumulate in the liver after immunization with live irradiation-attenuated P. berghei sporozoites. IFN-γ production by Plasmodium antigen-specific CD8+ T cells is dependent on functional Batf3. In addition, our results demonstrate that the dysfunctional cDC-CD8+ T-cell axis correlates with MHC class II upregulation on splenic CD8α- DCs. Collectively, these findings underscore the essential role of CD8α+ DCs in robust protection induced by experimental live-attenuated malaria vaccines.
Collapse
Affiliation(s)
- G N Montagna
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany.,Depto. de Microbiologia, Immunologia e Parasitologia, UNIFESP, Sao Paolo, Brazil
| | - A Biswas
- Institute of Medical Microbiology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - K Hildner
- University Hospital Erlangen, Medical Department 1, Erlangen, Germany
| | - K Matuschewski
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany.,Institute of Biology, Humboldt University, Berlin, Germany
| | - I R Dunay
- Institute of Medical Microbiology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| |
Collapse
|
42
|
Frevert U, Krzych U. Plasmodium cellular effector mechanisms and the hepatic microenvironment. Front Microbiol 2015; 6:482. [PMID: 26074888 PMCID: PMC4445044 DOI: 10.3389/fmicb.2015.00482] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/01/2015] [Indexed: 12/23/2022] Open
Abstract
Plasmodium falciparum malaria remains one of the most serious health problems globally. Immunization with attenuated parasites elicits multiple cellular effector mechanisms capable of eliminating Plasmodium liver stages. However, malaria liver stage (LS) immunity is complex and the mechanisms effector T cells use to locate the few infected hepatocytes in the large liver in order to kill the intracellular LS parasites remain a mystery to date. Here, we review our current knowledge on the behavior of CD8 effector T cells in the hepatic microvasculature, in malaria and other hepatic infections. Taking into account the unique immunological and lymphogenic properties of the liver, we discuss whether classical granule-mediated cytotoxicity might eliminate infected hepatocytes via direct cell contact or whether cytokines might operate without cell–cell contact and kill Plasmodium LSs at a distance. A thorough understanding of the cellular effector mechanisms that lead to parasite death hence sterile protection is a prerequisite for the development of a successful malaria vaccine to protect the 40% of the world’s population currently at risk of Plasmodium infection.
Collapse
Affiliation(s)
- Ute Frevert
- Division of Medical Parasitology, Department of Microbiology, New York University School of Medicine , New York, NY, USA
| | - Urszula Krzych
- Division of Malaria Vaccine Development, Department of Cellular Immunology, Walter Reed Army Institute of Research , Silver Spring, MD, USA
| |
Collapse
|
43
|
Whitacre DC, Espinosa DA, Peters CJ, Jones JE, Tucker AE, Peterson DL, Zavala FP, Milich DR. P. falciparum and P. vivax Epitope-Focused VLPs Elicit Sterile Immunity to Blood Stage Infections. PLoS One 2015; 10:e0124856. [PMID: 25933001 PMCID: PMC4416889 DOI: 10.1371/journal.pone.0124856] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 03/17/2015] [Indexed: 01/09/2023] Open
Abstract
In order to design P. falciparum preerythrocytic vaccine candidates, a library of circumsporozoite (CS) T and B cell epitopes displayed on the woodchuck hepatitis virus core antigen (WHcAg) VLP platform was produced. To test the protective efficacy of the WHcAg-CS VLPs, hybrid CS P. berghei/P. falciparum (Pb/Pf) sporozoites were used to challenge immunized mice. VLPs carrying 1 or 2 different CS repeat B cell epitopes and 3 VLPs carrying different CS non-repeat B cell epitopes elicited high levels of anti-insert antibodies (Abs). Whereas, VLPs carrying CS repeat B cell epitopes conferred 98% protection of the liver against a 10,000 Pb/Pf sporozoite challenge, VLPs carrying the CS non-repeat B cell eptiopes were minimally-to-non-protective. One-to-three CS-specific CD4/CD8 T cell sites were also fused to VLPs, which primed CS-specific as well as WHcAg-specific T cells. However, a VLP carrying only the 3 T cell domains failed to protect against a sporozoite challenge, indicating a requirement for anti-CS repeat Abs. A VLP carrying 2 CS repeat B cell epitopes and 3 CS T cell sites in alum adjuvant elicited high titer anti-CS Abs (endpoint dilution titer >1x106) and provided 80–100% protection against blood stage malaria. Using a similar strategy, VLPs were constructed carrying P. vivax CS repeat B cell epitopes (WHc-Pv-78), which elicited high levels of anti-CS Abs and conferred 99% protection of the liver against a 10,000 Pb/Pv sporozoite challenge and elicited sterile immunity to blood stage infection. These results indicate that immunization with epitope-focused VLPs carrying selected B and T cell epitopes from the P. falciparum and P. vivax CS proteins can elicit sterile immunity against blood stage malaria. Hybrid WHcAg-CS VLPs could provide the basis for a bivalent P. falciparum/P. vivax malaria vaccine.
Collapse
MESH Headings
- Animals
- Antibodies, Protozoan/immunology
- CD4-Positive T-Lymphocytes/immunology
- Epitopes, B-Lymphocyte/chemistry
- Epitopes, B-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/chemistry
- Epitopes, T-Lymphocyte/immunology
- Hepatitis B Virus, Woodchuck/immunology
- Immunity
- Immunization
- Life Cycle Stages
- Malaria, Falciparum/immunology
- Malaria, Falciparum/parasitology
- Malaria, Falciparum/prevention & control
- Malaria, Vivax/immunology
- Malaria, Vivax/parasitology
- Malaria, Vivax/prevention & control
- Mice, Inbred C57BL
- Plasmodium falciparum/immunology
- Plasmodium vivax/immunology
- Protozoan Proteins/immunology
- Rabbits
- Repetitive Sequences, Amino Acid
- Reproducibility of Results
- Virion/immunology
Collapse
Affiliation(s)
- David C. Whitacre
- Vaccine Research Institute of San Diego, San Diego, California, United States of America
- VLP Biotech, Inc., San Diego, California, United States of America
| | - Diego A. Espinosa
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Cory J. Peters
- Vaccine Research Institute of San Diego, San Diego, California, United States of America
- VLP Biotech, Inc., San Diego, California, United States of America
| | - Joyce E. Jones
- Vaccine Research Institute of San Diego, San Diego, California, United States of America
- VLP Biotech, Inc., San Diego, California, United States of America
| | - Amy E. Tucker
- VLP Biotech, Inc., San Diego, California, United States of America
| | - Darrell L. Peterson
- Department of Biochemistry, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Fidel P. Zavala
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - David R. Milich
- Vaccine Research Institute of San Diego, San Diego, California, United States of America
- VLP Biotech, Inc., San Diego, California, United States of America
- * E-mail:
| |
Collapse
|
44
|
Radtke AJ, Tse SW, Zavala F. From the draining lymph node to the liver: the induction and effector mechanisms of malaria-specific CD8+ T cells. Semin Immunopathol 2015; 37:211-20. [PMID: 25917387 PMCID: PMC5600878 DOI: 10.1007/s00281-015-0479-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 03/15/2015] [Indexed: 10/23/2022]
Abstract
Parasitic protozoa cause considerable disease in humans and, due to their intracellular life cycle, induce robust CD8(+) T cell responses. A greater understanding of the factors that promote and maintain CD8(+) T cell-mediated immunity against these pathogens is likely needed for the development of effective vaccines. Immunization with radiation-attenuated sporozoites, the infectious stage of the malaria parasite transmitted by mosquitoes, is an excellent model to study these questions as CD8(+) T cells specific for a single epitope can completely eliminate parasite infection in the liver. Furthermore, live, radiation-attenuated parasites represent the "gold standard" for malaria vaccination. Here, we will highlight recent studies aimed at understanding the factors required for the induction, recruitment, and maintenance of effector and memory CD8(+) T cells against malaria liver stages.
Collapse
Affiliation(s)
- Andrea J. Radtke
- Lymphocyte Biology Section, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Sze-Wah Tse
- Program in Cellular and Molecular Medicine of Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Fidel Zavala
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
45
|
Bijker EM, Nganou-Makamdop K, van Gemert GJ, Zavala F, Cockburn I, Sauerwein RW. Studying the effect of chloroquine on sporozoite-induced protection and immune responses in Plasmodium berghei malaria. Malar J 2015; 14:130. [PMID: 25889324 PMCID: PMC4389414 DOI: 10.1186/s12936-015-0626-2] [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: 12/24/2014] [Accepted: 02/20/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Sporozoite immunization of animals and humans under a chemo-prophylactic cover of chloroquine (CPS-CQ) efficiently induces sterile protection against malaria. In humans, CPS-CQ is strikingly more efficient than immunization with radiation attenuated sporozoites (RAS), raising the hypothesis that this might be partially due to CQ. Chloroquine, an established anti-malarial drug, is also well known for its immune modulating properties including improvement of cross-presentation. The aim of this study was to investigate whether co-administration of CQ during sporozoite immunization improves cellular responses and protective efficacy in Plasmodium berghei models. METHODS A number of experiments in selected complimentary P. berghei murine models in Balb/cByJ and C57BL/6j mice was performed. First, the effect of CQ administration on the induction of protection and immune responses by RAS immunization was studied. Next, the effect of CQ on the induction of circumsporozoite (CS) protein-specific CD8(+) T cells by immunization with P. berghei parasites expressing a mutant CS protein was investigated. Finally, a direct comparison of CPS-CQ to CPS with mefloquine (MQ), an anti-malarial with little known immune modulating effects, was performed. RESULTS When CQ was co-administered during immunization with graded numbers of RAS, this did not lead to an increase in frequencies of total memory CD8(+) T cells or CS protein-specific CD8(+) T cells. Also parasite-specific cytokine production and protection remained unaltered. Replacement of CQ by MQ for CPS immunization resulted in significantly reduced percentages of IFNγ producing memory T cells in the liver (p = 0.01), but similar protection. CONCLUSIONS This study does not provide evidence for a direct beneficial effect of CQ on the induction of sporozoite-induced immune responses and protection in P. berghei malaria models. Alternatively, the higher efficiency of CPS compared to RAS might be explained by an indirect effect of CQ through limiting blood-stage exposure after immunization or to increased antigen exposure and, therefore, improved breadth of the immune response.
Collapse
Affiliation(s)
- Else M Bijker
- Department of Medical Microbiology, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Krystelle Nganou-Makamdop
- Department of Medical Microbiology, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands. .,Current address: Vaccine Research Centre; National Institutes of Health, 40 Convent drive, Bethesda, MD, 20892, USA.
| | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Fidel Zavala
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, John Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA.
| | - Ian Cockburn
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, John Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA. .,Current address: John Curtin School of Medical Research, Australian National University, GPO Box 334, Canberra City, ACT 2600, Australia.
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
46
|
Espinosa DA, Gutierrez GM, Rojas-López M, Noe AR, Shi L, Tse SW, Sinnis P, Zavala F. Proteolytic Cleavage of the Plasmodium falciparum Circumsporozoite Protein Is a Target of Protective Antibodies. J Infect Dis 2015; 212:1111-9. [PMID: 25762791 DOI: 10.1093/infdis/jiv154] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 03/03/2015] [Indexed: 11/13/2022] Open
Abstract
Studies in animals and human volunteers demonstrate that antibodies against the repeat-region of the Plasmodium circumsporozoite protein (CSP) abrogate sporozoite infection. However, the realization that the N- and C- terminal regions flanking the repeats play essential roles in parasite infectivity raised the possibility that they could be targeted by protective antibodies. We characterized a monoclonal antibody (mAb5D5) specific for the N-terminus of the P. falciparum CSP, which inhibits the proteolytic cleavage of the CSP, a key requirement for parasite infection of hepatocytes. Adoptive transfer of mAb5D5 strongly inhibits the in vivo infection of sporozoites expressing the N-terminus of P. falciparum CSP, and this protection is greatly enhanced when combined with antirepeat antibodies. Our results show that antibodies interfering with molecular processes required for parasite infectivity can exert a strong in vivo protective activity and indicate that pre-erythrocytic vaccines against Plasmodium should include the CSP N-terminal region.
Collapse
Affiliation(s)
- Diego A Espinosa
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore
| | | | - Maricarmen Rojas-López
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore
| | | | - Lirong Shi
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore
| | - Sze-Wah Tse
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore
| | - Photini Sinnis
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore
| |
Collapse
|
47
|
Bertolino P, Bowen DG. Malaria and the liver: immunological hide-and-seek or subversion of immunity from within? Front Microbiol 2015; 6:41. [PMID: 25741320 PMCID: PMC4332352 DOI: 10.3389/fmicb.2015.00041] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 01/12/2015] [Indexed: 12/28/2022] Open
Abstract
During the pre-erythrocytic asymptomatic phase of malarial infection, sporozoites develop transiently inside less than 100 hepatocytes that subsequently release thousands of merozoites. Killing of these hepatocytes by cytotoxic T cells (CTLs) confers protection to subsequent malarial infection, suggesting that this bottleneck phase in the parasite life cycle can be targeted by vaccination. During natural transmission, although some CTLs are generated in the skin draining lymph nodes, they are unable to eliminate the parasite, suggesting that the liver is important for the sporozoite to escape immune surveillance. The contribution of the organ to this process is unclear. Based on the known ability of several hepatic antigen-presenting cells (APCs) to induce primary activation of CD8 T cells and tolerance, malarial antigens presented by both infected hepatocytes and/or hepatic cross-presenting APCs should result in tolerance. However, our latest model predicts that due to the low frequency of infected hepatocytes, some T cells recognizing sporozoite epitopes with high affinity should differentiate into CTLs. In this review, we discuss two possible models to explain why CTLs generated in the liver and skin draining lymph nodes are unable to eliminate the parasite: (1) sporozoites harness the tolerogenic property of the liver; (2) CTLs are not tolerized but fail to detect infected cells due to sparse infection of hepatocytes and the very short liver stage. We propose that while malaria sporozoites might use the ability of the liver to tolerize both naive and effector cells, they have also developed strategies to decrease the probability of encounter between CTLs and infected liver cells. Thus, we predict that to achieve protection, vaccination strategies should aim to boost intrahepatic activation and/or increase the chance of encounter between sporozoite-specific CTLs and infected hepatocytes.
Collapse
Affiliation(s)
- Patrick Bertolino
- Liver Immunology Group, Centenary Institute and AW Morrow Gastroenterology and Liver Centre, University of Sydney and Royal Prince Alfred Hospital Sydney, NSW, Australia
| | - David G Bowen
- Liver Immunology Group, Centenary Institute and AW Morrow Gastroenterology and Liver Centre, University of Sydney and Royal Prince Alfred Hospital Sydney, NSW, Australia
| |
Collapse
|
48
|
Radtke AJ, Kastenmüller W, Espinosa DA, Gerner MY, Tse SW, Sinnis P, Germain RN, Zavala FP, Cockburn IA. Lymph-node resident CD8α+ dendritic cells capture antigens from migratory malaria sporozoites and induce CD8+ T cell responses. PLoS Pathog 2015; 11:e1004637. [PMID: 25658939 PMCID: PMC4450069 DOI: 10.1371/journal.ppat.1004637] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 12/19/2014] [Indexed: 01/08/2023] Open
Abstract
Malaria infection begins when a female Anopheles mosquito injects Plasmodium sporozoites into the skin of its host during blood feeding. Skin-deposited sporozoites may enter the bloodstream and infect the liver, reside and develop in the skin, or migrate to the draining lymph nodes (DLNs). Importantly, the DLN is where protective CD8+ T cell responses against malaria liver stages are induced after a dermal route of infection. However, the significance of parasites in the skin and DLN to CD8+ T cell activation is largely unknown. In this study, we used genetically modified parasites, as well as antibody-mediated immobilization of sporozoites, to determine that active sporozoite migration to the DLNs is required for robust CD8+ T cell responses. Through dynamic in vivo and static imaging, we show the direct uptake of parasites by lymph-node resident DCs followed by CD8+ T cell-DC cluster formation, a surrogate for antigen presentation, in the DLNs. A few hours after sporozoite arrival to the DLNs, CD8+ T cells are primed by resident CD8α+ DCs with no apparent role for skin-derived DCs. Together, these results establish a critical role for lymph node resident CD8α+ DCs in CD8+ T cell priming to sporozoite antigens while emphasizing a requirement for motile sporozoites in the induction of CD8+ T cell-mediated immunity. Malaria is responsible for the deaths of 0.5–2 million people each year. A safe and effective vaccine is likely needed for the control or eradication of malaria. Immunization with irradiated sporozoites, the infectious stage of the parasite transmitted by mosquitoes, protects people against malaria through the activation of specialized effector cells called CD8+ T cells, which can eliminate live parasites. The induction of such malaria-specific CD8+ T cells is critically dependent on dendritic cells, a diverse population of antigen-presenting cells. It was previously unclear how dendritic cells acquire sporozoite antigens to induce the protective CD8+ T cell response. Using a combination of functional studies and high-resolution imaging, we report here that live sporozoites access skin-draining lymph nodes after infection and directly provide antigens to resident dendritic cells that in turn activate CD8+ T cells. These results underscore the importance of live, motile sporozoites in the induction of protective CD8+ T cell responses and provide a mechanistic understanding for the superior immunogenicity of whole parasite vaccines.
Collapse
Affiliation(s)
- Andrea J. Radtke
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Wolfgang Kastenmüller
- Lymphocyte Biology Section, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Diego A. Espinosa
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Michael Y. Gerner
- Lymphocyte Biology Section, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Sze-Wah Tse
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Photini Sinnis
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Ronald N. Germain
- Lymphocyte Biology Section, Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Fidel P. Zavala
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- * E-mail: (FPZ); (IAC)
| | - Ian A. Cockburn
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- * E-mail: (FPZ); (IAC)
| |
Collapse
|
49
|
Bayarsaikhan G, Akbari M, Yui K, Amino R. Antigen-driven focal inflammatory death of malaria liver stages. Front Microbiol 2015; 6:47. [PMID: 25699034 PMCID: PMC4316770 DOI: 10.3389/fmicb.2015.00047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 01/14/2015] [Indexed: 01/24/2023] Open
Abstract
Multiple immunizations using live irradiated sporozoites, the infectious plasmodial stage delivered into the host skin during a mosquito bite, can elicit sterile immunity to malaria. CD8+ T cells seem to play an essential role in this protective immunity, since their depletion consistently abolishes sterilizing protection in several experimental models. So far, only a few parasite antigens are known to induce CD8+ T cell-dependent protection, but none of them can reach the levels of protection afforded by live attenuated parasites. Systematic attempts to identify novel antigens associated with this efficient cellular protection were so far unsuccessful. In addition, the precise mechanisms involved in the recognition and elimination of parasitized hepatocytes in vivo by CD8+ T cells still remain obscure. Recently, it has been shown that specific effector CD8+ T cells, after recognition of parasitized hepatocytes, recruit specific and non-specific activated CD8+ T cells to the site of infection, resulting in the formation of cellular clusters around and in the further elimination of intracellular parasites. The significance of this finding is discussed in the perspective of a general mechanism of antigen-dependent focalized inflammation and its consequences for the elimination of malaria liver stages.
Collapse
Affiliation(s)
- Ganchimeg Bayarsaikhan
- Unité de Biologie et Génétique du Paludisme, Département Parasites et Insectes Vecteurs, Institut Pasteur , Paris, France ; Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University , Nagasaki, Japan
| | - Masoud Akbari
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University , Nagasaki, Japan
| | - Katsuyuki Yui
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University , Nagasaki, Japan
| | - Rogerio Amino
- Unité de Biologie et Génétique du Paludisme, Département Parasites et Insectes Vecteurs, Institut Pasteur , Paris, France
| |
Collapse
|
50
|
Crompton PD, Moebius J, Portugal S, Waisberg M, Hart G, Garver LS, Miller LH, Barillas-Mury C, Pierce SK. Malaria immunity in man and mosquito: insights into unsolved mysteries of a deadly infectious disease. Annu Rev Immunol 2014; 32:157-87. [PMID: 24655294 DOI: 10.1146/annurev-immunol-032713-120220] [Citation(s) in RCA: 225] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Malaria is a mosquito-borne disease caused by parasites of the obligate intracellular Apicomplexa phylum the most deadly of which, Plasmodium falciparum, prevails in Africa. Malaria imposes a huge health burden on the world's most vulnerable populations, claiming the lives of nearly one million children and pregnant women each year. Although there is keen interest in eradicating malaria, we do not yet have the necessary tools to meet this challenge, including an effective malaria vaccine and adequate vector control strategies. Here we review what is known about the mechanisms at play in immune resistance to malaria in both the human and mosquito hosts at each step in the parasite's complex life cycle with a view toward developing the tools that will contribute to the prevention of disease and death and, ultimately, to the goal of malaria eradication. In so doing, we hope to inspire immunologists to participate in defeating this devastating disease.
Collapse
|