1
|
Franco A, Flores-Garcia Y, Venezia J, Daoud A, Scott AL, Zavala F, Sullivan DJ. Hemozoin-induced IFN-γ production mediates innate immune protection against sporozoite infection. Microbes Infect 2024; 26:105343. [PMID: 38670216 DOI: 10.1016/j.micinf.2024.105343] [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: 10/16/2023] [Revised: 04/10/2024] [Accepted: 04/21/2024] [Indexed: 04/28/2024]
Abstract
Hemozoin is a crystal synthesized by Plasmodium parasites during hemoglobin digestion in the erythrocytic stage. The hemozoin released when the parasites egress from the red blood cell, which is complexed with parasite DNA, is cleared from the circulation by circulating and tissue-resident monocytes and macrophages, respectively. Recently, we reported that intravenous administration of purified hemozoin complexed with Plasmodium berghei DNA (HzPbDNA) resulted in an innate immune response that blocked liver stage development of sporozoites that was dose-dependent and time-limited. Here, we further characterize the organismal, cellular, and molecular events associated with this protective innate response in the liver and report that a large proportion of the IV administered HzPbDNA localized to F4/80+ cells in the liver and that the rapid and strong protection against liver-stage development waned quickly such that by 1 week post-HzPbDNA treatment animals were fully susceptible to infection. RNAseq of the liver after IV administration of HzPbDNA demonstrated that the rapid and robust induction of genes associated with the acute phase response, innate immune activation, cellular recruitment, and IFN-γ signaling observed at day 1 was largely absent at day 7. RNAseq analysis implicated NK cells as the major cellular source of IFN-γ. In vivo cell depletion and IFN-γ neutralization experiments supported the hypothesis that tissue-resident macrophages and NK cells are major contributors to the protective response and the NK cell-derived IFN-γ is key to induction of the mechanisms that block sporozoite development in the liver. These findings advance our understanding of the innate immune responses that prevent liver stage malaria infection.
Collapse
Affiliation(s)
- Adriano Franco
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, MD, 21205, USA
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, MD, 21205, USA
| | - Jarrett Venezia
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, MD, 21205, USA
| | - Abdel Daoud
- Department of Pathology, Johns Hopkins School of Medicine, 720 Rutland Avenue, Baltimore, MD, 21205, USA
| | - Alan L Scott
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, MD, 21205, USA
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, MD, 21205, USA
| | - David J Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, MD, 21205, USA.
| |
Collapse
|
2
|
Furtado R, Paul M, Zhang J, Sung J, Karell P, Kim RS, Caillat-Zucman S, Liang L, Felgner P, Bauleni A, Gama S, Buchwald A, Taylor T, Seydel K, Laufer M, Delahaye F, Daily JP, Lauvau G. Cytolytic circumsporozoite-specific memory CD4 + T cell clones are expanded during Plasmodium falciparum infection. Nat Commun 2023; 14:7726. [PMID: 38001069 PMCID: PMC10673885 DOI: 10.1038/s41467-023-43376-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Clinical immunity against Plasmodium falciparum infection develops in residents of malaria endemic regions, manifesting in reduced clinical symptoms during infection and in protection against severe disease but the mechanisms are not fully understood. Here, we compare the cellular and humoral immune response of clinically immune (0-1 episode over 18 months) and susceptible (at least 3 episodes) during a mild episode of Pf malaria infection in a malaria endemic region of Malawi, by analysing peripheral blood samples using high dimensional mass cytometry (CyTOF), spectral flow cytometry and single-cell transcriptomic analyses. In the clinically immune, we find increased proportions of circulating follicular helper T cells and classical monocytes, while the humoral immune response shows characteristic age-related differences in the protected. Presence of memory CD4+ T cell clones with a strong cytolytic ZEB2+ T helper 1 effector signature, sharing identical T cell receptor clonotypes and recognizing the Pf-derived circumsporozoite protein (CSP) antigen are found in the blood of the Pf-infected participants gaining protection. Moreover, in clinically protected participants, ZEB2+ memory CD4+ T cells express lower level of inhibitory and chemotactic receptors. We thus propose that clonally expanded ZEB2+ CSP-specific cytolytic memory CD4+ Th1 cells may contribute to clinical immunity against the sporozoite and liver-stage Pf malaria.
Collapse
Affiliation(s)
- Raquel Furtado
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
- RF: BioNTech US, 40 Erie Street, Cambridge, MA, 02139, USA
| | - Mahinder Paul
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Jinghang Zhang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Joowhan Sung
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Paul Karell
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Ryung S Kim
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Sophie Caillat-Zucman
- Université de Paris, AP-HP, Hôpital Saint-Louis, Laboratoire d'Immunologie et Histocompatiblité, INSERM UMR976, 75010, Paris, France
| | - Li Liang
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Philip Felgner
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Andy Bauleni
- Malaria Alert Centre, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Syze Gama
- Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Andrea Buchwald
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Terrie Taylor
- Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
- Department of Osteopathic Medical Specialties, Michigan State University, East Lansing, MI, 48824, USA
| | - Karl Seydel
- Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
- Department of Osteopathic Medical Specialties, Michigan State University, East Lansing, MI, 48824, USA
| | - Miriam Laufer
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Fabien Delahaye
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
- FD: Precision Oncology, Sanofi, Vitry sur Seine, France
| | - Johanna P Daily
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA.
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, 10461, USA.
| | - Grégoire Lauvau
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA.
| |
Collapse
|
3
|
Rajneesh, Tiwari R, Singh VK, Kumar A, Gupta RP, Singh AK, Gautam V, Kumar R. Advancements and Challenges in Developing Malaria Vaccines: Targeting Multiple Stages of the Parasite Life Cycle. ACS Infect Dis 2023; 9:1795-1814. [PMID: 37708228 DOI: 10.1021/acsinfecdis.3c00332] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Malaria, caused by Plasmodium species, remains a major global health concern, causing millions of deaths annually. While the introduction of the RTS,S vaccine has shown promise, there is a pressing need for more effective vaccines due to the emergence of drug-resistant parasites and insecticide-resistant vectors. However, the complex life cycle and genetic diversity of the parasite, technical obstacles, limited funding, and the impact of the 2019 pandemic have hindered progress in malaria vaccine development. This review focuses on advancements in malaria vaccine development, particularly the ongoing clinical trials targeting antigens from different stages of the Plasmodium life cycle. Additionally, we discuss the rationale, strategies, and challenges associated with vaccine design, aiming to enhance the immune response and protective efficacy of vaccine candidates. A cost-effective and multistage vaccine could hold the key to controlling and eradicating malaria.
Collapse
Affiliation(s)
- Rajneesh
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Rahul Tiwari
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Vishal K Singh
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Awnish Kumar
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Rohit P Gupta
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
- Department of Applied Microbiology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Akhilesh K Singh
- Faculty of Dental Science, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Vibhav Gautam
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Rajiv Kumar
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
4
|
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: 13] [Impact Index Per Article: 13.0] [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
|
5
|
Nakamae S, Miyagawa S, Ogawa K, Kamiya M, Taniguchi M, Ono A, Kawaguchi M, Teklemichael AA, Jian JY, Araki T, Katagami Y, Mukai H, Annoura T, Yui K, Hirayama K, Kawakami S, Mizukami S. Induction of liver-resident memory T cells and protection at liver-stage malaria by mRNA-containing lipid nanoparticles. Front Immunol 2023; 14:1116299. [PMID: 37680630 PMCID: PMC10482405 DOI: 10.3389/fimmu.2023.1116299] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 07/25/2023] [Indexed: 09/09/2023] Open
Abstract
Recent studies have suggested that CD8+ liver-resident memory T (TRM) cells are crucial in the protection against liver-stage malaria. We used liver-directed mRNA-containing lipid nanoparticles (mRNA-LNPs) to induce liver TRM cells in a murine model. Single-dose intravenous injections of ovalbumin mRNA-LNPs effectively induced antigen-specific cytotoxic T lymphocytes in a dose-dependent manner in the liver on day 7. TRM cells (CD8+ CD44hi CD62Llo CD69+ KLRG1-) were induced 5 weeks after immunization. To examine the protective efficacy, mice were intramuscularly immunized with two doses of circumsporozoite protein mRNA-LNPs at 3-week intervals and challenged with sporozoites of Plasmodium berghei ANKA. Sterile immunity was observed in some of the mice, and the other mice showed a delay in blood-stage development when compared with the control mice. mRNA-LNPs therefore induce memory CD8+ T cells that can protect against sporozoites during liver-stage malaria and may provide a basis for vaccines against the disease.
Collapse
Affiliation(s)
- Sayuri Nakamae
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Satoshi Miyagawa
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
- Laboratory for Drug Discovery and Disease Research, SHIONOGI & CO., LTD., Osaka, Japan
| | - Koki Ogawa
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Mariko Kamiya
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Mayumi Taniguchi
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Akari Ono
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Maho Kawaguchi
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Awet Alem Teklemichael
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Jiun-Yu Jian
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Tamasa Araki
- Department of Parasitology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Yukimi Katagami
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Hidefumi Mukai
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Takeshi Annoura
- Department of Parasitology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Katsuyuki Yui
- Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Kenji Hirayama
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Shigeru Kawakami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki, Japan
| | - Shusaku Mizukami
- Department of Immune Regulation, Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Nagasaki, Japan
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Nagasaki, Japan
| |
Collapse
|
6
|
Rajakaruna H, Ganusov VV. Mathematical Modeling to Guide Experimental Design: T Cell Clustering as a Case Study. Bull Math Biol 2022; 84:103. [PMID: 35978047 PMCID: PMC9548402 DOI: 10.1007/s11538-022-01063-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/28/2022] [Indexed: 11/02/2022]
Abstract
Mathematical modeling provides a rigorous way to quantify immunological processes and discriminate between alternative mechanisms driving specific biological phenomena. It is typical that mathematical models of immunological phenomena are developed by modelers to explain specific sets of experimental data after the data have been collected by experimental collaborators. Whether the available data are sufficient to accurately estimate model parameters or to discriminate between alternative models is not typically investigated. While previously collected data may be sufficient to guide development of alternative models and help estimating model parameters, such data often do not allow to discriminate between alternative models. As a case study, we develop a series of power analyses to determine optimal sample sizes that allow for accurate estimation of model parameters and for discrimination between alternative models describing clustering of CD8 T cells around Plasmodium liver stages. In our typical experiments, mice are infected intravenously with Plasmodium sporozoites that invade hepatocytes (liver cells), and then activated CD8 T cells are transferred into the infected mice. The number of T cells found in the vicinity of individual infected hepatocytes at different times after T cell transfer is counted using intravital microscopy. We previously developed a series of mathematical models aimed to explain highly variable number of T cells per parasite; one of such models, the density-dependent recruitment (DDR) model, fitted the data from preliminary experiments better than the alternative models, such as the density-independent exit (DIE) model. Here, we show that the ability to discriminate between these alternative models depends on the number of parasites imaged in the analysis; analysis of about [Formula: see text] parasites at 2, 4, and 8 h after T cell transfer will allow for over 95% probability to select the correct model. The type of data collected also has an impact; following T cell clustering around individual parasites over time (called as longitudinal (LT) data) allows for a more precise and less biased estimates of the parameters of the DDR model than that generated from a more traditional way of imaging individual parasites in different liver areas/mice (cross-sectional (CS) data). However, LT imaging comes at a cost of a need to keep the mice alive under the microscope for hours which may be ethically unacceptable. We finally show that the number of time points at which the measurements are taken also impacts the precision of estimation of DDR model parameters; in particular, measuring T cell clustering at one time point does not allow accurately estimating all parameters of the DDR model. Using our case study, we propose a general framework on how mathematical modeling can be used to guide experimental designs and power analyses of complex biological processes.
Collapse
Affiliation(s)
- Harshana Rajakaruna
- Battelle Center for Mathematical Medicine, The Abigail Wexner Research Institute, 575 Children's Crossroad, Columbus, OH, 43215, USA.
| | - Vitaly V Ganusov
- Department of Mathematics, University of Tennessee, Knoxville, TN, 37996, USA.
- Department of Microbiology, University of Tennessee, Knoxville, TN, 37996, USA.
| |
Collapse
|
7
|
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
|
8
|
Hon C, Friesen J, Ingmundson A, Scheppan D, Hafalla JCR, Müller K, Matuschewski K. Conservation of S20 as an Ineffective and Disposable IFNγ-Inducing Determinant of Plasmodium Sporozoites Indicates Diversion of Cellular Immunity. Front Microbiol 2021; 12:703804. [PMID: 34421862 PMCID: PMC8377727 DOI: 10.3389/fmicb.2021.703804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/07/2021] [Indexed: 11/19/2022] Open
Abstract
Despite many decades of research to develop a malaria vaccine, only one vaccine candidate has been explored in pivotal phase III clinical trials. This candidate subunit vaccine consists of a portion of a single Plasmodium antigen, circumsporozoite protein (CSP). This antigen was initially identified in the murine malaria model and shown to contain an immunodominant and protective CD8+ T cell epitope specific to the H-2Kd (BALB/c)-restricted genetic background. A high-content screen for CD8+ epitopes in the H2Kb/Db (C57BL/6)-restricted genetic background, identified two distinct dominant epitopes. In this study, we present a characterization of one corresponding antigen, the Plasmodium sporozoite-specific protein S20. Plasmodium berghei S20 knockout sporozoites and liver stages developed normally in vitro and in vivo. This potent infectivity of s20(-) sporozoites permitted comparative analysis of knockout and wild-type parasites in cell-based vaccination. Protective immunity of irradiation-arrested s20(-) sporozoites in single, double and triple immunizations was similar to irradiated unaltered sporozoites in homologous challenge experiments. These findings demonstrate the presence of an immunogenic Plasmodium pre-erythrocytic determinant, which is not essential for eliciting protection. Although S20 is not needed for colonization of the mammalian host and for initiation of a blood infection, it is conserved amongst Plasmodium species. Malarial parasites express conserved, immunogenic proteins that are not required to establish infection but might play potential roles in diverting cellular immune responses.
Collapse
Affiliation(s)
- Calvin Hon
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany
| | - Johannes Friesen
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany.,Medical Care Unit Labor 28 GmbH, Berlin, Germany
| | - Alyssa Ingmundson
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany.,Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Diana Scheppan
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Julius C R Hafalla
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Katja Müller
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany.,Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Kai Matuschewski
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany.,Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
9
|
Sahu T, Gehrke EJ, Flores-Garcia Y, Mlambo G, Romano JD, Coppens I. Chemoprophylaxis vaccination with a Plasmodium liver stage autophagy mutant affords enhanced and long-lasting protection. NPJ Vaccines 2021; 6:98. [PMID: 34376691 PMCID: PMC8355287 DOI: 10.1038/s41541-021-00360-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 07/06/2021] [Indexed: 11/09/2022] Open
Abstract
Genetically attenuated sporozoite vaccines can elicit long-lasting protection against malaria but pose risks of breakthrough infection. Chemoprophylaxis vaccination (CVac) has proven to be the most effective vaccine strategy against malaria. Here, we demonstrate that a liver stage-specific autophagy mutant of Plasmodium berghei (ATG8 overexpressor), when used as a live vaccine under a CVac regimen, provides superior long-lasting protection, in both inbred and outbred mice, as compared to WT-CVac. Uniquely, the protection elicited by this mutant is predominantly dependent on a CD8+ T-cell response through an IFN-γ-independent mechanism and is associated with a stable population of antigen-experienced CD8+ T cells. Jointly, our findings support the exploitation of liver-stage mutants as vaccines under a CVac protocol. This vaccination strategy is also a powerful model to study the mechanisms of protective immunity and discover new protective antigens.
Collapse
Affiliation(s)
- Tejram Sahu
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Ella J Gehrke
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Godfree Mlambo
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Julia D Romano
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins Malaria Research Institute, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
10
|
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: 1.3] [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
|
11
|
Li L, Zeng Z. Live Imaging of Innate and Adaptive Immune Responses in the Liver. Front Immunol 2020; 11:564768. [PMID: 33042143 PMCID: PMC7527534 DOI: 10.3389/fimmu.2020.564768] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/13/2020] [Indexed: 12/21/2022] Open
Abstract
Immune response in the liver is determined by the spatial organization and cellular dynamics of hepatic immune cells. The liver vasculature accommodates abundant tissue-resident innate immune cells, such as Kupffer cells, natural killer cells, and natural killer T cells, to ensure efficient intravascular immunosurveillance. The fenestrated sinusoids also allow direct contact between circulating T cells and non-canonical antigen-presenting cells, such as hepatocytes, to instruct adaptive immune responses. Distinct cellular behaviors are exploited by liver immune cells to exert proper functions. Intravital imaging enables real-time visualization of individual immune cell in living animals, representing a powerful tool in dissecting the spatiotemporal features of intrahepatic immune cells during steady state and liver diseases. This review summarizes current advances in liver immunology prompted by in vivo imaging, with a particular focus on liver-resident innate immune cells and hepatic T cells.
Collapse
Affiliation(s)
- Lu Li
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhutian Zeng
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
12
|
Phares TW, Kotraiah V, Karunarathne DS, Huang J, Browne CD, Buontempo P, Mansour M, Noe AR, Wykes MN, Pannucci J, Tsuji M, Gutierrez GM. A Peptide-Based PD1 Antagonist Enhances T-Cell Priming and Efficacy of a Prophylactic Malaria Vaccine and Promotes Survival in a Lethal Malaria Model. Front Immunol 2020; 11:1377. [PMID: 32733457 PMCID: PMC7363839 DOI: 10.3389/fimmu.2020.01377] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 05/29/2020] [Indexed: 12/14/2022] Open
Abstract
The blockade of programmed cell death-1 (PD1) and its ligand PDL1 has been proven to be a successful immunotherapy against several cancers. Similar to cancer, PD1 contributes to the establishment of several chronic infectious diseases, including malaria. While monoclonal antibodies (mAbs) targeting checkpoint receptors are revolutionary in cancer treatment, the immune-related adverse events (irAEs) may prevent their utilization in prophylactic and therapeutic treatments of infectious diseases. The irAEs are, in part, due to the prolonged half-life of mAbs resulting in prolonged activation of the immune system. As an alternative modality to mAbs, peptides represent a viable option because they possess a shorter pharmacokinetic half-life and offer more formulation and delivery options. Here, we report on a 22-amino acid immunomodulatory peptide, LD01, derived from a Bacillus bacteria. When combined prophylactically with an adenovirus-based or irradiated sporozoite-based malaria vaccine, LD01 significantly enhanced antigen-specific CD8+ T cell expansion. Therapeutically, LD01 treatment of mice infected with a lethal malaria strain resulted in survival that was associated with lower numbers of FOXP3+Tbet+CD4+ regulatory T cells. Taken together, our results demonstrate that LD01 is a potent immunomodulator that acts upon the adaptive immune system to stimulate T cell responses both prophylactically and therapeutically.
Collapse
Affiliation(s)
- Timothy W Phares
- Explorations in Global Health (ExGloH), Leidos Inc., Frederick, MD, United States
| | - Vinayaka Kotraiah
- Explorations in Global Health (ExGloH), Leidos Inc., Frederick, MD, United States
| | | | - Jing Huang
- The Aaron Diamond AIDS Research Center, New York, NY, United States
| | | | - Peter Buontempo
- Explorations in Global Health (ExGloH), Leidos Inc., Frederick, MD, United States
| | - Marc Mansour
- Explorations in Global Health (ExGloH), Leidos Inc., Frederick, MD, United States
| | - Amy R Noe
- Leidos Life Sciences, Leidos Inc., Frederick, MD, United States
| | | | - James Pannucci
- Explorations in Global Health (ExGloH), Leidos Inc., Frederick, MD, United States
| | - Moriya Tsuji
- The Aaron Diamond AIDS Research Center, New York, NY, United States
| | - Gabriel M Gutierrez
- Explorations in Global Health (ExGloH), Leidos Inc., Frederick, MD, United States
| |
Collapse
|
13
|
De Niz M, Carvalho T, Penha-Gonçalves C, Agop-Nersesian C. Intravital imaging of host-parasite interactions in organs of the thoracic and abdominopelvic cavities. Cell Microbiol 2020; 22:e13201. [PMID: 32149435 DOI: 10.1111/cmi.13201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/03/2020] [Accepted: 03/06/2020] [Indexed: 12/16/2022]
Abstract
Infections with protozoan and helminthic parasites affect multiple organs in the mammalian host. Imaging pathogens in their natural environment takes a more holistic view on biomedical aspects of parasitic infections. Here, we focus on selected organs of the thoracic and abdominopelvic cavities most commonly affected by parasites. Parasitic infections of these organs are often associated with severe medical complications or have health implications beyond the infected individual. Intravital imaging has provided a more dynamic picture of the host-parasite interplay and contributed not only to our understanding of the various disease pathologies, but has also provided fundamental insight into the biology of the parasites.
Collapse
Affiliation(s)
- Mariana De Niz
- Institute of Cell Biology, University of Bern, Bern, Switzerland.,Instituto de Medicina Molecular - João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Tânia Carvalho
- Instituto de Medicina Molecular - João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | | | | |
Collapse
|
14
|
Abstract
Immunity to malaria has been linked to the availability and function of helper CD4+ T cells, cytotoxic CD8+ T cells and γδ T cells that can respond to both the asymptomatic liver stage and the symptomatic blood stage of Plasmodium sp. infection. These T cell responses are also thought to be modulated by regulatory T cells. However, the precise mechanisms governing the development and function of Plasmodium-specific T cells and their capacity to form tissue-resident and long-lived memory populations are less well understood. The field has arrived at a point where the push for vaccines that exploit T cell-mediated immunity to malaria has made it imperative to define and reconcile the mechanisms that regulate the development and functions of Plasmodium-specific T cells. Here, we review our current understanding of the mechanisms by which T cell subsets orchestrate host resistance to Plasmodium infection on the basis of observational and mechanistic studies in humans, non-human primates and rodent models. We also examine the potential of new experimental strategies and human infection systems to inform a new generation of approaches to harness T cell responses against malaria.
Collapse
|
15
|
Imai T, Suzue K, Ngo-Thanh H, Ono S, Orita W, Suzuki H, Shimokawa C, Olia A, Obi S, Taniguchi T, Ishida H, Van Kaer L, Murata S, Tanaka K, Hisaeda H. Fluctuations of Spleen Cytokine and Blood Lactate, Importance of Cellular Immunity in Host Defense Against Blood Stage Malaria Plasmodium yoelii. Front Immunol 2019; 10:2207. [PMID: 31608052 PMCID: PMC6773889 DOI: 10.3389/fimmu.2019.02207] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/02/2019] [Indexed: 12/27/2022] Open
Abstract
Our previous studies of protective immunity and pathology against blood stage malaria parasites have shown that not only CD4+ T cells, but also CD8+ T cells and macrophages, are important for host defense against blood stage malaria infection. Furthermore, we found that Plasmodium yoelii 17XNL (PyNL) parasitizes erythroblasts, the red blood cell (RBC) precursor cells, which then express MHC class I molecules. In the present study, we analyzed spleen cytokine production. In CD8+ T cell-depleted mice, IL-10 production in early stage infection was increased over two-fold relative to infected control animals and IL-10+ CD3- cells were increased, whereas IFN-γ production in the late stage of infection was decreased. At day 16 after PyNL infection, CD8+ T cells produced more IFN-γ than CD4+ T cells. We evaluated the involvement of the immunoproteasome in induction of immune CD8+ T cells, and the role of Fas in protection against PyNL both of which are downstream of IFN-γ. In cell transfer experiments, at least the single molecules LMP7, LMP2, and PA28 are not essential for CD8+ T cell induction. The Fas mutant LPR mouse was weaker in resistance to PyNL infection than WT mice, and 20% of the animals died. LPR-derived parasitized erythroid cells exhibited less externalization of phosphatidylserine (PS), and phagocytosis by macrophages was impaired. Furthermore, we tried to identify the cause of death in malaria infection. Blood lactate concentration was increased in the CD8+ T cell-depleted PyNL-infected group at day 19 (around peak parasitemia) to similar levels as day 7 after infection with a lethal strain of Py. When we injected mice with lactate at day 4 and 6 of PyNL infection, all mice died at day 8 despite demonstrating low parasitemia, suggesting that hyperlactatemia is one of the causes of death in CD8+ T cell-depleted PyNL-infected mice. We conclude that CD8+ T cells might control cytokine production to some extent and regulate hyperparasitemia and hyperlactatemia in protection against blood stage malaria parasites.
Collapse
Affiliation(s)
- Takashi Imai
- Department of Infectious Diseases and Host Defense, Gunma University Graduate School of Medicine, Maebashi, Japan.,Department of Parasitology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazutomo Suzue
- Department of Infectious Diseases and Host Defense, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Ha Ngo-Thanh
- Department of Infectious Diseases and Host Defense, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Suguri Ono
- Department of Infectious Diseases and Host Defense, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Wakako Orita
- Department of Infectious Diseases and Host Defense, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Haruka Suzuki
- Department of Infectious Diseases and Host Defense, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Chikako Shimokawa
- Department of Infectious Diseases and Host Defense, Gunma University Graduate School of Medicine, Maebashi, Japan.,Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Alex Olia
- Department of Infectious Diseases and Host Defense, Gunma University Graduate School of Medicine, Maebashi, Japan.,Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Seiji Obi
- Department of Infectious Diseases and Host Defense, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Tomoyo Taniguchi
- Center for Medical Education, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Hidekazu Ishida
- Department of Parasitology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Shigeo Murata
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Keiji Tanaka
- Laboratory of Protein Metabolism, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hajime Hisaeda
- Department of Parasitology, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
16
|
Kelemen RK, Rajakaruna H, Cockburn IA, Ganusov VV. Clustering of Activated CD8 T Cells Around Malaria-Infected Hepatocytes Is Rapid and Is Driven by Antigen-Specific Cells. Front Immunol 2019; 10:2153. [PMID: 31616407 PMCID: PMC6764016 DOI: 10.3389/fimmu.2019.02153] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/28/2019] [Indexed: 01/23/2023] Open
Abstract
Malaria, a disease caused by parasites of the Plasmodium genus, begins when Plasmodium-infected mosquitoes inject malaria sporozoites while searching for blood. Sporozoites migrate from the skin via blood to the liver, infect hepatocytes, and form liver stages which in mice 48 h later escape into blood and cause clinical malaria. Vaccine-induced activated or memory CD8 T cells are capable of locating and eliminating all liver stages in 48 h, thus preventing the blood-stage disease. However, the rules of how CD8 T cells are able to locate all liver stages within a relatively short time period remains poorly understood. We recently reported formation of clusters consisting of variable numbers of activated CD8 T cells around Plasmodium yoelii (Py)-infected hepatocytes. Using a combination of experimental data and mathematical models we now provide additional insights into mechanisms of formation of these clusters. First, we show that a model in which cluster formation is driven exclusively by T-cell-extrinsic factors, such as variability in "attractiveness" of different liver stages, cannot explain distribution of cluster sizes in different experimental conditions. In contrast, the model in which cluster formation is driven by the positive feedback loop (i.e., larger clusters attract more CD8 T cells) can accurately explain the available data. Second, while both Py-specific CD8 T cells and T cells of irrelevant specificity (non-specific CD8 T cells) are attracted to the clusters, we found no evidence that non-specific CD8 T cells play a role in cluster formation. Third and finally, mathematical modeling suggested that formation of clusters occurs rapidly, within few hours after adoptive transfer of CD8 T cells, thus illustrating high efficiency of CD8 T cells in locating their targets in complex peripheral organs, such as the liver. Taken together, our analysis provides novel insights into and attempts to discriminate between alternative mechanisms driving the formation of clusters of antigen-specific CD8 T cells in the liver.
Collapse
Affiliation(s)
- Reka K. Kelemen
- Institute of Science and Technology, Vienna, Austria
- Genome Science and Technology Program, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Harshana Rajakaruna
- Department of Microbiology, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Ian A. Cockburn
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Vitaly V. Ganusov
- Genome Science and Technology Program, University of Tennessee, Knoxville, Knoxville, TN, United States
- Department of Microbiology, University of Tennessee, Knoxville, Knoxville, TN, United States
| |
Collapse
|
17
|
Tedla MG, Every AL, Scheerlinck JPY. Investigating immune responses to parasites using transgenesis. Parasit Vectors 2019; 12:303. [PMID: 31202271 PMCID: PMC6570953 DOI: 10.1186/s13071-019-3550-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 06/03/2019] [Indexed: 11/10/2022] Open
Abstract
Parasites comprise diverse and complex organisms, which substantially impact human and animal health. Most parasites have complex life-cycles, and by virtue of co-evolution have developed multifaceted, often life-cycle stage-specific relationships with the immune system of their hosts. The complexity in the biology of many parasites often limits our knowledge of parasite-specific immune responses, to in vitro studies only. The relatively recent development of methods to stably manipulate the genetic make-up of many parasites has allowed a better understanding of host-parasite interactions, particularly in vivo. In this regard, the use of transgenic parasites can facilitate the study of immunomodulatory mechanisms under in vivo conditions. Therefore, in this review, we specifically highlighted the current developments in the use of transgenic parasites to unravel the host's immune response to different life-cycle stages of some key parasite species such as Leishmania, Schistosoma, Toxoplasma, Plasmodium and Trypanosome and to some degree, the use of transgenic nematode parasites is also briefly discussed.
Collapse
Affiliation(s)
- Mebrahtu G. Tedla
- Centre for Animal Biotechnology, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Melbourne, VIC 3010 Australia
| | - Alison L. Every
- Centre for Animal Biotechnology, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Melbourne, VIC 3010 Australia
- Present Address: College of Science, Health and Engineering, La Trobe University, Melbourne, VIC 3086 Australia
| | - Jean-Pierre Y. Scheerlinck
- Centre for Animal Biotechnology, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Melbourne, VIC 3010 Australia
| |
Collapse
|
18
|
Moormann AM, Nixon CE, Forconi CS. Immune effector mechanisms in malaria: An update focusing on human immunity. Parasite Immunol 2019; 41:e12628. [PMID: 30972776 DOI: 10.1111/pim.12628] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 04/02/2019] [Accepted: 04/08/2019] [Indexed: 12/12/2022]
Abstract
The past decade has witnessed dramatic decreases in malaria-associated mortality and morbidity around the world. This progress has largely been due to intensified malaria control measures, implementation of rapid diagnostics and establishing a network to anticipate and mitigate antimalarial drug resistance. However, the ultimate tool for malaria prevention is the development and implementation of an effective vaccine. To date, malaria vaccine efforts have focused on determining which of the thousands of antigens expressed by Plasmodium falciparum are instrumental targets of protective immunity. The antigenic variation and antigenic polymorphisms arising in parasite genes under immune selection present a daunting challenge for target antigen selection and prioritization, and is a given caveat when interpreting immune recall responses or results from monovalent vaccine trials. Other immune evasion strategies executed by the parasite highlight the myriad of ways in which it can become a recurrent infection. This review provides an update on immune effector mechanisms in malaria and focuses on our improved ability to interrogate the complexity of human immune system, accelerated by recent methodological advances. Appreciating how the human immune landscape influences the effectiveness and longevity of antimalarial immunity will help explain which conditions are necessary for immune effector mechanisms to prevail.
Collapse
Affiliation(s)
- Ann M Moormann
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Christina E Nixon
- Department of Pathology and Lab Medicine, Brown University, Providence, Rhode Island
| | - Catherine S Forconi
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| |
Collapse
|
19
|
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: 2.8] [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
|
20
|
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: 119] [Impact Index Per Article: 19.8] [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
|
21
|
CD8+ T Cell Activation Leads to Constitutive Formation of Liver Tissue-Resident Memory T Cells that Seed a Large and Flexible Niche in the Liver. Cell Rep 2018; 25:68-79.e4. [DOI: 10.1016/j.celrep.2018.08.094] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 07/23/2018] [Accepted: 08/30/2018] [Indexed: 01/27/2023] Open
|
22
|
Gola A, Silman D, Walters AA, Sridhar S, Uderhardt S, Salman AM, Halbroth BR, Bellamy D, Bowyer G, Powlson J, Baker M, Venkatraman N, Poulton I, Berrie E, Roberts R, Lawrie AM, Angus B, Khan SM, Janse CJ, Ewer KJ, Germain RN, Spencer AJ, Hill AVS. Prime and target immunization protects against liver-stage malaria in mice. Sci Transl Med 2018; 10:10/460/eaap9128. [DOI: 10.1126/scitranslmed.aap9128] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 03/08/2018] [Accepted: 08/20/2018] [Indexed: 12/24/2022]
Abstract
Despite recent advances in treatment and vector control, malaria is still a leading cause of death, emphasizing the need for an effective vaccine. The malaria life cycle can be subdivided into three stages: the invasion and growth within liver hepatocytes (pre-erythrocytic stage), the blood stage (erythrocytic stage), and, finally, the sexual stage (occurring within the mosquito vector). Antigen (Ag)-specific CD8+ T cells are effectively induced by heterologous prime-boost viral vector immunization and known to correlate with liver-stage protection. However, liver-stage malaria vaccines have struggled to generate and maintain the high numbers of Plasmodium-specific circulating T cells necessary to confer sterile protection. We describe an alternative “prime and target” vaccination strategy aimed specifically at inducing high numbers of tissue-resident memory T cells present in the liver at the time of hepatic infection. This approach bypasses the need for very high numbers of circulating T cells and markedly increases the efficacy of subunit immunization against liver-stage malaria with clinically relevant Ags and clinically tested viral vectors in murine challenge models. Translation to clinical use has begun, with encouraging results from a pilot safety and feasibility trial of intravenous chimpanzee adenovirus vaccination in humans. This work highlights the value of a prime-target approach for immunization against malaria and suggests that this strategy may represent a more general approach for prophylaxis or immunotherapy of other liver infections and diseases.
Collapse
|
23
|
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: 2.5] [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
|
24
|
Nlinwe ON, Kusi KA, Adu B, Sedegah M. T-cell responses against Malaria: Effect of parasite antigen diversity and relevance for vaccine development. Vaccine 2018; 36:2237-2242. [PMID: 29573877 DOI: 10.1016/j.vaccine.2018.03.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 01/21/2018] [Accepted: 03/07/2018] [Indexed: 11/24/2022]
Abstract
The on-going agenda for global malaria elimination will require the development of additional disease control and prevention measures since currently available tools are showing signs of inadequacy. Malaria vaccines are seen as one such important addition to the control arsenal since vaccines have proven to be highly effective public health tools against important human diseases. Both cell-mediated and antibody responses are generally believed to be important for malaria parasite control, although the exact targets of T and B cell responses against malaria have not been clearly defined. However, our current understanding of the immune response to malaria suggests that T cell responses against multiple antigenic targets may potentially be key for the development of a highly efficacious malaria vaccine. This review takes a comprehensive look at the available literature on T cell-mediated immunity against all human stages of the malaria parasite and the effect of antigen diversity on these responses. The implications of these interrelationships for the development of an effective vaccine for malaria are also highlighted.
Collapse
Affiliation(s)
- Omarine Nfor Nlinwe
- Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P.O. Box LG 581, Legon, Accra, Ghana.
| | - Kwadwo Asamoah Kusi
- Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P.O. Box LG 581, Legon, Accra, Ghana.
| | - Bright Adu
- Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, P.O. Box LG 581, Legon, Accra, Ghana.
| | - Martha Sedegah
- Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD 209 l0-7500, USA.
| |
Collapse
|
25
|
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: 1.1] [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
|
26
|
Moris P, Jongert E, van der Most RG. Characterization of T-cell immune responses in clinical trials of the candidate RTS,S malaria vaccine. Hum Vaccin Immunother 2017; 14:17-27. [PMID: 28934066 PMCID: PMC5791571 DOI: 10.1080/21645515.2017.1381809] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The candidate malaria vaccine RTS,S has demonstrated 45.7% efficacy over 18 months against all clinical disease in a phase-III field study of African children. RTS,S targets the circumsporozoite protein (CSP), which is expressed on the Plasmodium sporozoite during the pre-erythrocyte stage of its life-cycle; the stage between mosquito bite and liver infection. Early in the development of RTS,S, it was recognized that CSP-specific cell-mediated immunity (CMI) was required to complement CSP-specific antibody-mediated immunity. In reviewing RTS,S clinical studies, associations between protection and various types of CMI (CSP-specific CD4+ T cells and INF-γ ELISPOTs) have been identified, but not consistently. It is plausible that certain CD4+ T cells support antibody responses or co-operate with other immune-cell types to potentially elicit protection. However, the identities of vaccine correlates of protection, implicating either CSP-specific antibodies or T cells remain elusive, suggesting that RTS,S clinical trials may benefit from additional immunogenicity analyses that can be informed by the results of controlled human malaria infection studies.
Collapse
|
27
|
Sacci JB, Hollingdale MR, Sedegah M. Cellular immune response to DNA and vaccinia prime-boost immunization kills Plasmodium yoelii-infected hepatocytes in vitro. Pathog Dis 2017; 75:3798571. [PMID: 28475711 DOI: 10.1093/femspd/ftx051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 05/03/2017] [Indexed: 11/13/2022] Open
Abstract
Background Plasmid DNA encoding Plasmodium yoelii circumsporozoite protein (PyCSP) followed by boosting with recombinant vaccinia virus containing the PyCSP elicited significant protective immunity in mice that was primarily mediated by CD8+ T-cell responses directed to P. yoelii -infected hepatocytes. This study was to further explore protection using in vitro cultures of P. yoelii parasites in mouse hepatocytes. Spleen cells from DNA/vaccinia virus-immunized mice were co-cultured in vitro with mouse hepatocytes containing developing P. yoelii liver stage parasites. A semipermeable membrane separating spleen cells and hepatocytes was used to demonstrate if cell-to-cell contact was required. Inhibitors of mediators likely involved in spleen cell killing were added to these co-cultures. Spleen cells from immunized mice inhibited in vitro P. yoelii parasite development, and inhibition was eliminated by separating effectors and targets with the semipermeable membrane. Additionally, inhibitors of inducible nitric oxide synthase, caspase activation, NF-κB activation as well as antibodies against interferon-gamma (IFN-γ) and ICAM-1 reduced parasite inhibition. These findings suggest that direct contact between spleen cells from immunized mice and P. yoelii-infected hepatocytes is required for eliminating liver stage parasites and provide more insight into CD8+ T-cell-mediated inhibition of malaria liver stage development.
Collapse
Affiliation(s)
- John B Sacci
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | - Martha Sedegah
- Malaria Program, Naval Medical Research Center, Silver Spring, MD 20910, USA
| |
Collapse
|
28
|
Kreutzfeld O, Müller K, Matuschewski K. Engineering of Genetically Arrested Parasites (GAPs) For a Precision Malaria Vaccine. Front Cell Infect Microbiol 2017; 7:198. [PMID: 28620583 PMCID: PMC5450620 DOI: 10.3389/fcimb.2017.00198] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 05/04/2017] [Indexed: 01/08/2023] Open
Abstract
Continuous stage conversion and swift changes in the antigenic repertoire in response to acquired immunity are hallmarks of complex eukaryotic pathogens, including Plasmodium species, the causative agents of malaria. Efficient elimination of Plasmodium liver stages prior to blood infection is one of the most promising malaria vaccine strategies. Here, we describe different genetically arrested parasites (GAPs) that have been engineered in Plasmodium berghei, P. yoelii and P. falciparum and compare their vaccine potential. A better understanding of the immunological mechanisms of prime and boost by arrested sporozoites and experimental strategies to enhance vaccine efficacy by further engineering existing GAPs into a more immunogenic form hold promise for continuous improvements of GAP-based vaccines. A critical hurdle for vaccines that elicit long-lasting protection against malaria, such as GAPs, is safety and efficacy in vulnerable populations. Vaccine research should focus on solutions toward turning malaria into a vaccine-preventable disease, which would offer an exciting new path of malaria control.
Collapse
Affiliation(s)
- Oriana Kreutzfeld
- Department of Molecular Parasitology, Institute of Biology, Humboldt UniversityBerlin, Germany
| | - Katja Müller
- Department of Molecular Parasitology, Institute of Biology, Humboldt UniversityBerlin, Germany
| | - Kai Matuschewski
- Department of Molecular Parasitology, Institute of Biology, Humboldt UniversityBerlin, Germany
| |
Collapse
|
29
|
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: 2.4] [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
|
30
|
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: 2.0] [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
|
31
|
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: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 01/18/2016] [Indexed: 10/22/2022]
|
32
|
Santana-Jorge KTO, Santos TM, Tartaglia NR, Aguiar EL, Souza RFS, Mariutti RB, Eberle RJ, Arni RK, Portela RW, Meyer R, Azevedo V. Putative virulence factors of Corynebacterium pseudotuberculosis FRC41: vaccine potential and protein expression. Microb Cell Fact 2016; 15:83. [PMID: 27184574 PMCID: PMC4869379 DOI: 10.1186/s12934-016-0479-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 05/03/2016] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Corynebacterium pseudotuberculosis, a facultative intracellular bacterial pathogen, is the etiological agent of caseous lymphadenitis (CLA), an infectious disease that affects sheep and goats and it is responsible for significant economic losses. The disease is characterized mainly by bacteria-induced caseous necrosis in lymphatic glands. New vaccines are needed for reliable control and management of CLA. Thus, the putative virulence factors SpaC, SodC, NanH, and PknG from C. pseudotuberculosis FRC41 may represent new target proteins for vaccine development and pathogenicity studies. RESULTS SpaC, PknG and NanH presented better vaccine potential than SodC after in silico analyses. A total of 136 B and T cell epitopes were predicted from the four putative virulence factors. A cluster analysis was performed to evaluate the redundancy degree among the sequences of the predicted epitopes; 57 clusters were formed, most of them (34) were single clusters. Two clusters from PknG and one from SpaC grouped epitopes for B and T-cell (MHC I and II). These epitopes can thus potentially stimulate a complete immune response (humoral and cellular) against C. pseudotuberculosis. Several other clusters, including two from NanH, grouped B-cell epitopes with either MHC I or II epitopes. The four target proteins were expressed in Escherichia coli. A purification protocol was developed for PknG expression. CONCLUSIONS In silico analyses show that the putative virulence factors SpaC, PknG and NanH present good potential for CLA vaccine development. Target proteins were successfully expressed in E. coli. A protocol for PknG purification is described.
Collapse
Affiliation(s)
- Karina T. O. Santana-Jorge
- />Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antonio Carlos, 6627, Pampulha, Belo Horizonte, 31270-901 Brazil
| | - Túlio M. Santos
- />Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antonio Carlos, 6627, Pampulha, Belo Horizonte, 31270-901 Brazil
- />Uniclon Biotecnologia, Belo Horizonte, MG Brazil
| | - Natayme R. Tartaglia
- />Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antonio Carlos, 6627, Pampulha, Belo Horizonte, 31270-901 Brazil
| | - Edgar L. Aguiar
- />Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antonio Carlos, 6627, Pampulha, Belo Horizonte, 31270-901 Brazil
| | - Renata F. S. Souza
- />Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antonio Carlos, 6627, Pampulha, Belo Horizonte, 31270-901 Brazil
| | - Ricardo B. Mariutti
- />Multiuser Center for Biomolecular Innovation, Instituto de Biociências, Letras e Ciências Exatas, Universidade Estadual Paulista “Júlio de Mesquita Filho”, São José Do Rio Preto, SP Brazil
| | - Raphael J. Eberle
- />Multiuser Center for Biomolecular Innovation, Instituto de Biociências, Letras e Ciências Exatas, Universidade Estadual Paulista “Júlio de Mesquita Filho”, São José Do Rio Preto, SP Brazil
| | - Raghuvir K. Arni
- />Multiuser Center for Biomolecular Innovation, Instituto de Biociências, Letras e Ciências Exatas, Universidade Estadual Paulista “Júlio de Mesquita Filho”, São José Do Rio Preto, SP Brazil
| | - Ricardo W. Portela
- />Laboratório de Imunologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, BA Brazil
| | - Roberto Meyer
- />Laboratório de Imunologia e Biologia Molecular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, BA Brazil
| | - Vasco Azevedo
- />Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antonio Carlos, 6627, Pampulha, Belo Horizonte, 31270-901 Brazil
| |
Collapse
|
33
|
Akbari M, Kimura K, Houts JT, Yui K. Intravital imaging of the immune responses during liver-stage malaria infection: An improved approach for fixing the liver. Parasitol Int 2016; 65:502-505. [PMID: 26921520 DOI: 10.1016/j.parint.2016.02.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 02/10/2016] [Accepted: 02/23/2016] [Indexed: 11/26/2022]
Abstract
The host-parasite relationship is one of the main themes of modern parasitology. Recent revolutions in science, including the development of various fluorescent proteins/probes and two-photon microscopy, have made it possible to directly visualize and study the mechanisms underlying the interaction between the host and pathogen. Here, we describe our method of preparing and setting-up the liver for our experimental approach of using intravital imaging to examine the interaction between Plasmodium berghei ANKA and antigen-specific CD8+ T cells during the liver-stage of the infection in four dimensions. Since the liver is positioned near the diaphragm, neutralization of respiratory movements is critical during the imaging process. In addition, blood circulation and temperature can be affected by the surgical exposure due to the anatomy and tissue structure of the liver. To control respiration, we recommend anesthesia with isoflurane inhalation at 1% during the surgery. In addition, our protocol introduces a cushion of gauze around the liver to avoid external pressure on the liver during intravital imaging using an inverted microscope, which makes it possible to image the liver tissue for long periods with minimal reduction in the blood circulation and with minimal displacement and tissue damage. The key point of this method is to reduce respiratory movements and external pressure on the liver tissue during intravital imaging.
Collapse
Affiliation(s)
- Masoud Akbari
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan.
| | - Kazumi Kimura
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan.
| | - James T Houts
- Patterson Scientific, 1160 Chess Drive Suite #9, Foster City, CA 94404, USA.
| | - Katsuyuki Yui
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan.
| |
Collapse
|
34
|
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: 0.9] [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
|
35
|
Marques PE, Oliveira AG, Chang L, Paula-Neto HA, Menezes GB. Understanding liver immunology using intravital microscopy. J Hepatol 2015; 63:733-42. [PMID: 26055800 DOI: 10.1016/j.jhep.2015.05.027] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/21/2015] [Accepted: 05/21/2015] [Indexed: 12/15/2022]
Abstract
The liver has come a long way since it was considered only a metabolic organ attached to the gastrointestinal tract. The simultaneous ascension of immunology and intravital microscopy evidenced the liver as a central axis in the immune system, controlling immune responses to local and systemic agents as well as disease tolerance. The multiple hepatic cell populations are organized in a vascular environment that promotes intimate cellular interactions, including initiation of innate and adaptive immune responses, rapid leukocyte recruitment, pathogen clearance and production of a variety of immune mediators. In this review, we focus on the advances in liver immunology supported by intravital microscopy in diseases such as isquemia/reperfusion, acute liver injury and infections.
Collapse
Affiliation(s)
- Pedro Elias Marques
- Laboratório de Imunobiofotônica, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Brazil
| | - André Gustavo Oliveira
- Laboratório de Imunobiofotônica, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Brazil
| | | | - Heitor Affonso Paula-Neto
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Brazil
| | - Gustavo Batista Menezes
- Laboratório de Imunobiofotônica, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Brazil.
| |
Collapse
|
36
|
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: 2.6] [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
|
37
|
de Koning-Ward TF, Gilson PR, Crabb BS. Advances in molecular genetic systems in malaria. Nat Rev Microbiol 2015; 13:373-87. [DOI: 10.1038/nrmicro3450] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
38
|
Imai T, Ishida H, Suzue K, Taniguchi T, Okada H, Shimokawa C, Hisaeda H. Cytotoxic activities of CD8⁺ T cells collaborate with macrophages to protect against blood-stage murine malaria. eLife 2015; 4. [PMID: 25760084 PMCID: PMC4366679 DOI: 10.7554/elife.04232] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 02/24/2015] [Indexed: 12/14/2022] Open
Abstract
The protective immunity afforded by CD8+ T cells against blood-stage malaria remains controversial because no MHC class I molecules are displayed on parasite-infected human erythrocytes. We recently reported that rodent malaria parasites infect erythroblasts that express major histocompatibility complex (MHC) class I antigens, which are recognized by CD8+ T cells. In this study, we demonstrate that the cytotoxic activity of CD8+ T cells contributes to the protection of mice against blood-stage malaria in a Fas ligand (FasL)-dependent manner. Erythroblasts infected with malarial parasites express the death receptor Fas. CD8+ T cells induce the externalization of phosphatidylserine (PS) on the infected erythroblasts in a cell-to-cell contact-dependent manner. PS enhances the engulfment of the infected erythroid cells by phagocytes. As a PS receptor, T-cell immunoglobulin-domain and mucin-domain-containing molecule 4 (Tim-4) contributes to the phagocytosis of malaria-parasite-infected cells. Our findings provide insight into the molecular mechanisms underlying the protective immunity exerted by CD8+ T cells in collaboration with phagocytes. DOI:http://dx.doi.org/10.7554/eLife.04232.001 The immune system consists of several different types of cell that work together to prevent infection and disease. For example, immune cells called cytotoxic CD8+ T cells kill tumor cells or other cells that are infected. To do so, the CD8+ T cells must recognize certain molecules on the surface of the tumor or infected cells and bind to them. Malaria is an infectious disease caused by the Plasmodium parasite, which is transferred between individuals by mosquitoes. The parasite is able to evade the immune system—so much so that it is not well understood how the immune system tries to respond to stop the infection. This has made it difficult to develop a vaccine that protects against malaria. During the latter stages of a malaria infection, the parasite infects the host's red blood cells. It was long believed that CD8+ T cells did not help to eliminate the red blood cells that had been infected by Plasmodium. However, recent work in mice suggested that CD8+ T cells do respond to infected erythroblasts—precursor cells that develop into red blood cells—and that CD8+ T cells help protect mice against blood-stage malaria. Now, Imai et al. describe how the CD8+ T cells in mice help to kill erythroblasts infected with Plasmodium yoelli, a species of the parasite used to study malaria in mice. The infected cells display a protein called Fas on their surface. Imai et al. found that, during a malaria infection, the CD8+ T cells produce a protein that can interact with Fas. This interaction causes the infected cell to move a signaling molecule to its outside surface, which encourages another type of immune cell to engulf and destroy the infected cell. This knowledge of how CD8+ T cells fight Plasmodium parasites in the bloodstream could now help to develop new types of blood-stage vaccine for malaria. DOI:http://dx.doi.org/10.7554/eLife.04232.002
Collapse
Affiliation(s)
- Takashi Imai
- Department of Parasitology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hidekazu Ishida
- Microbiological Research Institute, Otsuka Pharmaceutical Co., Ltd, Tokushima, Japan
| | - Kazutomo Suzue
- Department of Parasitology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Tomoyo Taniguchi
- Department of Parasitology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hiroko Okada
- Department of Parasitology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Chikako Shimokawa
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Science, Yokohama, Japan
| | - Hajime Hisaeda
- Department of Parasitology, Gunma University Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
39
|
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: 0.8] [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
|
40
|
The subcellular location of ovalbumin in Plasmodium berghei blood stages influences the magnitude of T-cell responses. Infect Immun 2014; 82:4654-65. [PMID: 25156724 DOI: 10.1128/iai.01940-14] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Model antigens are frequently introduced into pathogens to study determinants that influence T-cell responses to infections. To address whether an antigen's subcellular location influences the nature and magnitude of antigen-specific T-cell responses, we generated Plasmodium berghei parasites expressing the model antigen ovalbumin (OVA) either in the parasite cytoplasm or on the parasitophorous vacuole membrane (PVM). For cytosolic expression, OVA alone or conjugated to mCherry was expressed from a strong constitutive promoter (OVAhsp70 or OVA::mCherryhsp70); for PVM expression, OVA was fused to HEP17/EXP1 (OVA::Hep17hep17). Unexpectedly, OVA expression in OVAhsp70 parasites was very low, but when OVA was fused to mCherry (OVA::mCherryhsp70), it was highly expressed. OVA expression in OVA::Hep17hep17 parasites was strong but significantly less than that in OVA::mCherryhsp70 parasites. These transgenic parasites were used to examine the effects of antigen subcellular location and expression level on the development of T-cell responses during blood-stage infections. While all OVA-expressing parasites induced activation and proliferation of OVA-specific CD8(+) T cells (OT-I) and CD4(+) T cells (OT-II), the level of activation varied: OVA::Hep17hep17 parasites induced significantly stronger splenic and intracerebral OT-I and OT-II responses than those of OVA::mCherryhsp70 parasites, but OVA::mCherryhsp70 parasites promoted stronger OT-I and OT-II responses than those of OVAhsp70 parasites. Despite lower OVA expression levels, OVA::Hep17hep17 parasites induced stronger T-cell responses than those of OVA::mCherryhsp70 parasites. These results indicate that unconjugated cytosolic OVA is not stably expressed in Plasmodium parasites and, importantly, that its cellular location and expression level influence both the induction and magnitude of parasite-specific T-cell responses. These parasites represent useful tools for studying the development and function of antigen-specific T-cell responses during malaria infection.
Collapse
|
41
|
Abstract
Protective immunity against preerythrocytic malaria parasite infection is difficult to achieve. Intracellular Plasmodium parasites likely minimize antigen presentation by surface-expressed major histocompatibility complex class I (MHC-I) molecules on infected cells, yet they actively remodel their host cells by export of parasite factors. Whether exported liver-stage proteins constitute better candidates for MHC-I antigen presentation to CD8+ T lymphocytes remains unknown. Here, we systematically characterized the contribution of protein export to the magnitude of antigen-specific T-cell responses against Plasmodium berghei liver-stage parasites in C57BL/6 mice. We generated transgenic sporozoites that secrete a truncated ovalbumin (OVA) surrogate antigen only in the presence of an amino-terminal protein export element. Immunization with live attenuated transgenic sporozoites revealed that antigen export was not critical for CD8+ T-cell priming but enhanced CD8+ T-cell proliferation in the liver. Upon transfer of antigen-specific CD8+ T cells, liver-stage parasites secreting the target protein were eliminated more efficiently. We conclude that Plasmodium parasites strictly control protein export during liver infection to minimize immune recognition. Strategies that enhance the discharge of parasite proteins into infected hepatocytes could improve the efficacy of candidate preerythrocytic malaria vaccines. Vaccine development against Plasmodium parasites remains a priority in malaria research. The most advanced malaria subunit vaccine candidates contain Plasmodium surface proteins with important roles for parasite vital functions. A fundamental question is whether recognition by effector CD8+ T cells is restricted to sporozoite surface antigens or extends to parasite proteins that are synthesized during the extensive parasite expansion phase in the liver. Using a surrogate model antigen, we found that a cytoplasmic antigen is able to induce robust protective CD8+ T-cell responses, but protein export further enhances immunogenicity and protection. Our results show that a cytoplasmic localization does not exclude a protein’s candidacy for malaria subunit vaccines and that protein secretion can enhance protective immunity.
Collapse
|
42
|
Mac-Daniel L, Buckwalter MR, Berthet M, Virk Y, Yui K, Albert ML, Gueirard P, Ménard R. Local immune response to injection of Plasmodium sporozoites into the skin. THE JOURNAL OF IMMUNOLOGY 2014; 193:1246-57. [PMID: 24981449 DOI: 10.4049/jimmunol.1302669] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Malarial infection is initiated when the sporozoite form of the Plasmodium parasite is inoculated into the skin by a mosquito. Sporozoites invade hepatocytes in the liver and develop into the erythrocyte-infecting form of the parasite, the cause of clinical blood infection. Protection against parasite development in the liver can be induced by injection of live attenuated parasites that do not develop in the liver and thus do not cause blood infection. Radiation-attenuated sporozoites (RAS) and genetically attenuated parasites are now considered as lead candidates for vaccination of humans against malaria. Although the skin appears as the preferable administration route, most studies in rodents, which have served as model systems, have been performed after i.v. injection of attenuated sporozoites. In this study, we analyzed the early response to Plasmodium berghei RAS or wild-type sporozoites (WTS) injected intradermally into C57BL/6 mice. We show that RAS have a similar in vivo distribution to WTS and that both induce a similar inflammatory response consisting of a biphasic recruitment of polymorphonuclear neutrophils and inflammatory monocytes in the skin injection site and proximal draining lymph node (dLN). Both WTS and RAS associate with neutrophils and resident myeloid cells in the skin and the dLN, transform inside CD11b(+) cells, and induce a Th1 cytokine profile in the dLN. WTS and RAS are also similarly capable of priming parasite-specific CD8(+) T cells. These studies delineate the early and local response to sporozoite injection into the skin, and suggest that WTS and RAS prime the host immune system in a similar fashion.
Collapse
Affiliation(s)
- Laura Mac-Daniel
- Unité de Biologie et Génétique du Paludisme, Institut Pasteur, 75724 Paris Cedex 15, France
| | - Matthew R Buckwalter
- Unité d'Immunobiologie des Cellules Dendritiques, Institut Pasteur, 75724 Paris Cedex 15, France; and
| | - Michèle Berthet
- Unité de Biologie et Génétique du Paludisme, Institut Pasteur, 75724 Paris Cedex 15, France
| | - Yasemin Virk
- Unité d'Immunobiologie des Cellules Dendritiques, Institut Pasteur, 75724 Paris Cedex 15, France; and
| | - Katsuyuki Yui
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Matthew L Albert
- Unité d'Immunobiologie des Cellules Dendritiques, Institut Pasteur, 75724 Paris Cedex 15, France; and
| | - Pascale Gueirard
- Unité de Biologie et Génétique du Paludisme, Institut Pasteur, 75724 Paris Cedex 15, France
| | - Robert Ménard
- Unité de Biologie et Génétique du Paludisme, Institut Pasteur, 75724 Paris Cedex 15, France;
| |
Collapse
|
43
|
Bijker EM, Teirlinck AC, Schats R, van Gemert GJ, van de Vegte-Bolmer M, van Lieshout L, IntHout J, Hermsen CC, Scholzen A, Visser LG, Sauerwein RW. Cytotoxic markers associate with protection against malaria in human volunteers immunized with Plasmodium falciparum sporozoites. J Infect Dis 2014; 210:1605-15. [PMID: 24872326 PMCID: PMC4208622 DOI: 10.1093/infdis/jiu293] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Immunization of healthy volunteers by bites from Plasmodium falciparum-infected mosquitoes during chloroquine chemoprophylaxis (hereafter, chemoprophylaxis and sporozoites [CPS] immunization) induces sterile protection against malaria. CPS-induced protection is mediated by immunity against pre-erythrocytic stages, presumably at least partially by cytotoxic cellular responses. We therefore aimed to investigate the association of CPS-induced cytotoxic T-cell markers with protection. METHODS In a double-blind randomized controlled trial, we performed dose titration of CPS immunization followed by homologous challenge infection in 29 subjects. Immune responses were assessed by in vitro restimulation of peripheral blood mononuclear cells and flow cytometry. RESULTS Dose-dependent complete protection was obtained in 4 of 5 volunteers after immunization with bites from 45 P. falciparum-infected mosquitoes, in 8 of 9 volunteers with bites from 30, and in 5 of 10 volunteers with bites from 15 (odds ratio [OR], 5.0; 95% confidence interval [CI], 1.5-17). Completely protected subjects had significantly higher proportions of CD4 T cells expressing the degranulation marker CD107a (OR, 8.4; 95% CI, 1.5-123; P = .011) and CD8 cells producing granzyme B (OR, 11; 95% CI, 1.9-212; P = .004) after P. falciparum restimulation. CONCLUSIONS These data underline the efficiency of CPS immunization to induce sterile protection and support a possible role for cytotoxic CD4 and CD8 T-cell responses in pre-erythrocytic immunity. CLINICAL TRIALS REGISTRATION NCT01218893.
Collapse
Affiliation(s)
| | | | | | | | | | - Lisette van Lieshout
- Department of Medical Microbiology, Department of Parasitology, Leiden University Medical Center, The Netherlands
| | - Joanna IntHout
- Department for Health Evidence, Section Biostatistics, Radboud university medical center, Nijmegen
| | | | | | | | | |
Collapse
|
44
|
Ingmundson A, Alano P, Matuschewski K, Silvestrini F. Feeling at home from arrival to departure: protein export and host cell remodelling during Plasmodium liver stage and gametocyte maturation. Cell Microbiol 2014; 16:324-33. [PMID: 24330249 DOI: 10.1111/cmi.12251] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/09/2013] [Accepted: 12/09/2013] [Indexed: 12/19/2022]
Abstract
Obligate intracellular pathogens actively remodel their host cells to boost propagation, survival, and persistence. Plasmodium falciparum, the causative agent of the most severe form of malaria, assembles a complex secretory system in erythrocytes. Export of parasite factors to the erythrocyte membrane is essential for parasite sequestration from the blood circulation and a major factor for clinical complications in falciparum malaria. Historic and recent molecular reports show that host cell remodelling is not exclusive to P. falciparum and that parasite-induced intra-erythrocytic membrane structures and protein export occur in several Plasmodia. Comparative analyses of P. falciparum asexual and sexual blood stages and imaging of liver stages from transgenic murine Plasmodium species show that protein export occurs in all intracellular phases from liver infection to sexual differentiation, indicating that mammalian Plasmodium species evolved efficient strategies to renovate erythrocytes and hepatocytes according to the specific needs of each life cycle phase. While the repertoireof identified exported proteins is remarkably expanded in asexual P. falciparum blood stages, the putative export machinery and known targeting signatures are shared across life cycle stages. A better understanding of the molecular mechanisms underlying Plasmodium protein export could assist in designing novel strategies to interrupt transmission between Anopheles mosquitoes and humans.
Collapse
Affiliation(s)
- Alyssa Ingmundson
- Max Planck Institute for Infection Biology, Parasitology Unit, 10117, Berlin, Germany
| | | | | | | |
Collapse
|
45
|
CD8+ T cells eliminate liver-stage Plasmodium berghei parasites without detectable bystander effect. Infect Immun 2014; 82:1460-4. [PMID: 24421043 DOI: 10.1128/iai.01500-13] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Immunization with attenuated Plasmodium sporozoites or viral vectored vaccines can induce protective CD8(+) T cells that can find and eliminate liver-stage malaria parasites. A key question is whether CD8(+) T cells must recognize and eliminate each parasite in the liver or whether bystander killing can occur. To test this, we transferred antigen-specific effector CD8(+) T cells to mice that were then coinfected with two Plasmodium berghei strains, only one of which could be recognized directly by the transferred T cells. We found that the noncognate parasites developed normally in these mice, demonstrating that bystander killing of parasites does not occur during the CD8(+) T cell response to malaria parasites. Rather, elimination of infected parasites is likely mediated by direct recognition of infected hepatocytes by antigen-specific CD8(+) T cells.
Collapse
|