1
|
Lee HJ, Moreira ML, Li S, Asatsuma T, Williams CG, Skinner OP, Asad S, Bramhall M, Jiang Z, Liu Z, Kerr AS, Engel JA, Soon MSF, Straube J, Barrera I, Murray E, Chen F, Nideffer J, Jagannathan P, Haque A. CD4 + T cells display a spectrum of recall dynamics during re-infection with malaria parasites. Nat Commun 2024; 15:5497. [PMID: 38944658 PMCID: PMC11214622 DOI: 10.1038/s41467-024-49879-6] [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: 08/08/2023] [Accepted: 06/24/2024] [Indexed: 07/01/2024] Open
Abstract
Children in malaria-endemic regions can experience repeated Plasmodium infections over short periods of time. Effects of re-infection on multiple co-existing CD4+ T cell subsets remain unresolved. Here, we examine antigen-experienced CD4+ T cells during re-infection in mice, using scRNA-seq/TCR-seq and spatial transcriptomics. TCR transgenic TEM cells initiate rapid Th1/Tr1 recall responses prior to proliferating, while GC Tfh counterparts are refractory, with TCM/Tfh-like cells exhibiting modest non-proliferative responses. Th1-recall is a partial facsimile of primary Th1-responses, with no upregulated effector-associated genes being unique to recall. Polyclonal, TCR-diverse, CD4+ T cells exhibit similar recall dynamics, with individual clones giving rise to multiple effectors including highly proliferative Th1/Tr1 cells, as well as GC Tfh and Tfh-like cells lacking proliferative capacity. Thus, we show substantial diversity in recall responses mounted by multiple co-existing CD4+ T cell subsets in the spleen, and present graphical user interfaces for studying gene expression dynamics and clonal relationships during re-infection.
Collapse
Affiliation(s)
- Hyun Jae Lee
- Department of Microbiology and Immunology, University of Melbourne, located at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Marcela L Moreira
- Department of Microbiology and Immunology, University of Melbourne, located at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Shihan Li
- Department of Microbiology and Immunology, University of Melbourne, located at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Takahiro Asatsuma
- Department of Microbiology and Immunology, University of Melbourne, located at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Cameron G Williams
- Department of Microbiology and Immunology, University of Melbourne, located at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Oliver P Skinner
- Department of Microbiology and Immunology, University of Melbourne, located at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Saba Asad
- Department of Microbiology and Immunology, University of Melbourne, located at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Michael Bramhall
- Department of Microbiology and Immunology, University of Melbourne, located at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Zhe Jiang
- Department of Microbiology and Immunology, University of Melbourne, located at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Zihan Liu
- Department of Microbiology and Immunology, University of Melbourne, located at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Ashlyn S Kerr
- Department of Microbiology and Immunology, University of Melbourne, located at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia
| | - Jessica A Engel
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD, Australia
| | - Megan S F Soon
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD, Australia
| | - Jasmin Straube
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD, Australia
- University of Queensland, Brisbane, QLD, Australia
| | | | - Evan Murray
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Fei Chen
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | | | - Prasanna Jagannathan
- Department of Medicine, Stanford University, CA, USA
- Department of Microbiology and Immunology, Stanford University, CA, USA
| | - Ashraful Haque
- Department of Microbiology and Immunology, University of Melbourne, located at The Peter Doherty Institute for Infection and Immunity, Parkville, VIC, Australia.
| |
Collapse
|
2
|
John L, Vijay R. Role of TAM Receptors in Antimalarial Humoral Immune Response. Pathogens 2024; 13:298. [PMID: 38668253 PMCID: PMC11054553 DOI: 10.3390/pathogens13040298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 03/29/2024] [Accepted: 03/31/2024] [Indexed: 04/29/2024] Open
Abstract
Immune response against malaria and the clearance of Plasmodium parasite relies on germinal-center-derived B cell responses that are temporally and histologically layered. Despite a well-orchestrated germinal center response, anti-Plasmodium immune response seldom offers sterilizing immunity. Recent studies report that certain pathophysiological features of malaria such as extensive hemolysis, hypoxia as well as the extrafollicular accumulation of short-lived plasmablasts may contribute to this suboptimal immune response. In this review, we summarize some of those studies and attempt to connect certain host intrinsic features in response to the malarial disease and the resultant gaps in the immune response.
Collapse
Affiliation(s)
- Lijo John
- Department of Veterinary Biochemistry, Kerala Veterinary and Animal Sciences University, Pookode 673576, Kerala, India
- Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60047, USA
| | - Rahul Vijay
- Center for Cancer Cell Biology, Immunology and Infection, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60047, USA
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60047, USA
| |
Collapse
|
3
|
Lansink LIM, Skinner OP, Engel JA, Lee HJ, Soon MSF, Williams CG, SheelaNair A, Pernold CPS, Laohamonthonkul P, Akter J, Stoll T, Hill MM, Talman AM, Russell A, Lawniczak M, Jia X, Chua B, Anderson D, Creek DJ, Davenport MP, Khoury DS, Haque A. Systemic host inflammation induces stage-specific transcriptomic modification and slower maturation in malaria parasites. mBio 2023; 14:e0112923. [PMID: 37449844 PMCID: PMC10470790 DOI: 10.1128/mbio.01129-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 06/02/2023] [Indexed: 07/18/2023] Open
Abstract
Maturation rates of malaria parasites within red blood cells (RBCs) can be influenced by host nutrient status and circadian rhythm; whether host inflammatory responses can also influence maturation remains less clear. Here, we observed that systemic host inflammation induced in mice by an innate immune stimulus, lipopolysaccharide (LPS), or by ongoing acute Plasmodium infection, slowed the progression of a single cohort of parasites from one generation of RBC to the next. Importantly, plasma from LPS-conditioned or acutely infected mice directly inhibited parasite maturation during in vitro culture, which was not rescued by supplementation, suggesting the emergence of inhibitory factors in plasma. Metabolomic assessments confirmed substantial alterations to the plasma of LPS-conditioned and acutely infected mice, and identified a small number of candidate inhibitory metabolites. Finally, we confirmed rapid parasite responses to systemic host inflammation in vivo using parasite scRNA-seq, noting broad impairment in transcriptional activity and translational capacity specifically in trophozoites but not rings or schizonts. Thus, we provide evidence that systemic host inflammation rapidly triggered transcriptional alterations in circulating blood-stage Plasmodium trophozoites and predict candidate inhibitory metabolites in the plasma that may impair parasite maturation in vivo. IMPORTANCE Malaria parasites cyclically invade, multiply, and burst out of red blood cells. We found that a strong inflammatory response can cause changes to the composition of host plasma, which directly slows down parasite maturation. Thus, our work highlights a new mechanism that limits malaria parasite growth in the bloodstream.
Collapse
Affiliation(s)
- Lianne I. M. Lansink
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
- Department of Biology, University of York, Wentworth Way, York, Yorkshire, United Kingdom
| | - Oliver P. Skinner
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | - Jessica A. Engel
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Hyun Jae Lee
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | - Megan S. F. Soon
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Cameron G. Williams
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | - Arya SheelaNair
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Clara P. S. Pernold
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | | | - Jasmin Akter
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Thomas Stoll
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Michelle M. Hill
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Arthur M. Talman
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, United Kingdom
- MIVEGEC, University of Montpellier, IRD, CNRS, Montpellier, France
| | - Andrew Russell
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, United Kingdom
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Mara Lawniczak
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, United Kingdom
| | - Xiaoxiao Jia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | - Brendon Chua
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | - Dovile Anderson
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Darren J. Creek
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Miles P. Davenport
- The Kirby Institute, University of New South Wales, Kensington, Sydney, New South Wales, Australia
| | - David S. Khoury
- The Kirby Institute, University of New South Wales, Kensington, Sydney, New South Wales, Australia
| | - Ashraful Haque
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
4
|
Ren Z, Shi Q, Xu S, Xu J, Yin Y, Lin Z, Xu S, Ma X, Liu Y, Zhu G, He X, Lu J, Li Y, Zhang W, Liu J, Yang Y, Han ET, Cao J, Lu F. Elicitation of T-cell-derived IFN-γ-dependent immunity by highly conserved Plasmodium ovale curtisi Duffy binding protein domain region II (PocDBP-RII). Parasit Vectors 2023; 16:269. [PMID: 37553591 PMCID: PMC10410920 DOI: 10.1186/s13071-023-05897-9] [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: 05/24/2023] [Accepted: 07/27/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Infections with Plasmodium ovale are widely distributed but rarely investigated, and the resulting burden of disease has been underestimated. Plasmodium ovale curtisi Duffy binding protein domain region II (PocDBP-RII) is an essential ligand for reticulocyte recognition and host cell invasion by P. ovale curtisi. However, the genomic variation, antigenicity and immunogenicity of PocDBP-RII remain major knowledge gaps. METHODS A total of 93 P. ovale curtisi samples were collected from migrant workers who returned to China from 17 countries in Africa between 2012 and 2016. The genetic polymorphism, natural selection and copy number variation (CNV) were investigated by sequencing and real-time PCR. The antigenicity and immunogenicity of the recombinant PocDBP-RII (rPocDBP-RII) protein were further examined, and the humoral and cellular responses of immunized mice were assessed using protein microarrays and flow cytometry. RESULTS Efficiently expressed and purified rPocDBP-RII (39 kDa) was successfully used as an antigen for immunization in mice. The haplotype diversity (Hd) of PocDBP-RII gene was 0.105, and the nucleotide diversity index (π) was 0.00011. No increased copy number was found among the collected isolates of P. ovale curtisi. Furthermore, rPocDBP-RII induced persistent antigen-specific antibody production with a serum IgG antibody titer of 1: 16,000. IFN-γ-producing T cells, rather than IL-10-producing cells, were activated in response to the stimulation of rPocDBP-RII. Compared to PBS-immunized mice (negative control), there was a higher percentage of CD4+CD44highCD62L- T cells (effector memory T cells) and CD8+CD44highCD62L+ T cells (central memory T cells) in rPocDBP-RII‑immunized mice. CONCLUSIONS PocDBP-RII sequences were highly conserved in clinical isolates of P. ovale curtisi. rPocDBP-RII protein could mediate protective blood-stage immunity through IFN-γ-producing CD4+ and CD8+ T cells and memory T cells, in addition to inducing specific antibodies. Our results suggested that rPocDBP-RII protein has potential as a vaccine candidate to provide assessment and guidance for malaria control and elimination activities.
Collapse
Affiliation(s)
- Zhenyu Ren
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Qiyang Shi
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory On Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Simin Xu
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
- Changshu Second People's Hospital, Suzhou, 215500, Jiangsu, People's Republic of China
| | - Jiahui Xu
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Yi Yin
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Zhijie Lin
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Sui Xu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory On Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Xiaoqin Ma
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory On Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Yaobao Liu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory On Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Guoding Zhu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory On Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Xinlong He
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Jingyuan Lu
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Yinyue Li
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Wenwen Zhang
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Jiali Liu
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Yun Yang
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Jun Cao
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory On Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China.
| | - Feng Lu
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.
- Affiliated Hospital of Yangzhou University, Yangzhou, 225000, People's Republic of China.
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.
| |
Collapse
|
5
|
Abstract
INTRODUCTION : Eradication of malaria remains one of the main aims of medicine. Despite progress in malaria treatment, mortality rate remains high, especially in the poorest parts of the world. Therefore, prevention through vaccines is fundamental and recent approval of the first effective vaccine reinforced this assumption. However, since the parasite cycle is complex, being composed of three stages, different types of vaccine targeting stage-specific antigens shall be developed. Moreover, the beneficial effect on vaccinated subjects can be tuned using compositions targeting different disease stages. AREA COVERED : We analysed the malaria vaccine patent landscape describing the most significant patents published after 2016, classified according to the different parasite stages targeted focusing on selected protein antigens or epitopes. We searched "malaria vaccine" on Patentscope and Espacenet. EXPERT OPINION : Pre-erythrocytic vaccines were boosted by RTS,S approval, but its partial efficacy, limited to sporozoites, calls for compositions active against other disease stages. In particular, multi-antigens vaccines could be more effective than single-stage ones, as they would activate an immune response more similar to that acquired in endemic regions. Furthermore, vaccine storage is another factor to be taken into account given the climate of the areas where malaria is widespread. More advanced technologies can lead to more effective and safer vaccines.
Collapse
Affiliation(s)
- Michael Quagliata
- Interdepartmental Research Unit of Peptide and Protein Chemistry and Biology, Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 13, I-50019 Sesto Fiorentino, Italy
| | - Anna Maria Papini
- Interdepartmental Research Unit of Peptide and Protein Chemistry and Biology, Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 13, I-50019 Sesto Fiorentino, Italy
| | - Paolo Rovero
- Interdepartmental Research Unit of Peptide and Protein Chemistry and Biology, Department of NeuroFarBa, University of Florence, Via Ugo Schiff 6, I-50019 Sesto Fiorentino, Italy
| |
Collapse
|
6
|
Chandley P, Ranjan R, Kumar S, Rohatgi S. Host-parasite interactions during Plasmodium infection: Implications for immunotherapies. Front Immunol 2023; 13:1091961. [PMID: 36685595 PMCID: PMC9845897 DOI: 10.3389/fimmu.2022.1091961] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Malaria is a global infectious disease that remains a leading cause of morbidity and mortality in the developing world. Multiple environmental and host and parasite factors govern the clinical outcomes of malaria. The host immune response against the Plasmodium parasite is heterogenous and stage-specific both in the human host and mosquito vector. The Plasmodium parasite virulence is predominantly associated with its ability to evade the host's immune response. Despite the availability of drug-based therapies, Plasmodium parasites can acquire drug resistance due to high antigenic variations and allelic polymorphisms. The lack of licensed vaccines against Plasmodium infection necessitates the development of effective, safe and successful therapeutics. To design an effective vaccine, it is important to study the immune evasion strategies and stage-specific Plasmodium proteins, which are targets of the host immune response. This review provides an overview of the host immune defense mechanisms and parasite immune evasion strategies during Plasmodium infection. Furthermore, we also summarize and discuss the current progress in various anti-malarial vaccine approaches, along with antibody-based therapy involving monoclonal antibodies, and research advancements in host-directed therapy, which can together open new avenues for developing novel immunotherapies against malaria infection and transmission.
Collapse
Affiliation(s)
- Pankaj Chandley
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India
| | - Ravikant Ranjan
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India
| | - Sudhir Kumar
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Soma Rohatgi
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India,*Correspondence: Soma Rohatgi,
| |
Collapse
|
7
|
Olatunde AC, Cornwall DH, Roedel M, Lamb TJ. Mouse Models for Unravelling Immunology of Blood Stage Malaria. Vaccines (Basel) 2022; 10:1525. [PMID: 36146602 PMCID: PMC9501382 DOI: 10.3390/vaccines10091525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Malaria comprises a spectrum of disease syndromes and the immune system is a major participant in malarial disease. This is particularly true in relation to the immune responses elicited against blood stages of Plasmodium-parasites that are responsible for the pathogenesis of infection. Mouse models of malaria are commonly used to dissect the immune mechanisms underlying disease. While no single mouse model of Plasmodium infection completely recapitulates all the features of malaria in humans, collectively the existing models are invaluable for defining the events that lead to the immunopathogenesis of malaria. Here we review the different mouse models of Plasmodium infection that are available, and highlight some of the main contributions these models have made with regards to identifying immune mechanisms of parasite control and the immunopathogenesis of malaria.
Collapse
Affiliation(s)
| | | | | | - Tracey J. Lamb
- Department of Pathology, University of Utah, Emma Eccles Jones Medical Research Building, 15 N Medical Drive E, Room 1420A, Salt Lake City, UT 84112, USA
| |
Collapse
|
8
|
SheelaNair A, Romanczuk AS, Aogo RA, Haldar RN, Lansink LIM, Cromer D, Salinas YG, Guy RK, McCarthy JS, Davenport MP, Haque A, Khoury DS. Similarly efficacious anti-malarial drugs SJ733 and pyronaridine differ in their ability to remove circulating parasites in mice. Malar J 2022; 21:49. [PMID: 35172826 PMCID: PMC8848794 DOI: 10.1186/s12936-022-04075-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/06/2022] [Indexed: 11/13/2022] Open
Abstract
Background Artemisinin-based combination therapy (ACT) has been a mainstay for malaria prevention and treatment. However, emergence of drug resistance has incentivised development of new drugs. Defining the kinetics with which circulating parasitized red blood cells (pRBC) are lost after drug treatment, referred to as the “parasite clearance curve”, has been critical for assessing drug efficacy; yet underlying mechanisms remain partly unresolved. The clearance curve may be shaped both by the rate at which drugs kill parasites, and the rate at which drug-affected parasites are removed from circulation. Methods In this context, two anti-malarials, SJ733, and an ACT partner drug, pyronaridine were compared against sodium artesunate in mice infected with Plasmodium berghei (strain ANKA). To measure each compound’s capacity for pRBC removal in vivo, flow cytometric monitoring of a single cohort of fluorescently-labelled pRBC was employed, and combined with ex vivo parasite culture to assess parasite maturation and replication. Results These three compounds were found to be similarly efficacious in controlling established infection by reducing overall parasitaemia. While sodium artesunate acted relatively consistently across the life-stages, single-dose SJ733 elicited a biphasic effect, triggering rapid, partly phagocyte-dependent removal of trophozoites and schizonts, followed by arrest of residual ring-stages. In contrast, pyronaridine abrogated maturation of younger parasites, with less pronounced effects on mature parasites, while modestly increasing pRBC removal. Conclusions Anti-malarials SJ733 and pyronaridine, though similarly efficacious in reducing overall parasitaemia in mice, differed markedly in their capacity to arrest replication and remove pRBC from circulation. Thus, similar parasite clearance curves can result for anti-malarials with distinct capacities to inhibit, kill and clear parasites.
Collapse
Affiliation(s)
- Arya SheelaNair
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, QLD, 4006, Australia
| | - Aleksandra S Romanczuk
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, QLD, 4006, Australia
| | - Rosemary A Aogo
- Infection Analytics Program, Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, 2052, Australia
| | - Rohit Nemai Haldar
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, QLD, 4006, Australia
| | - Lianne I M Lansink
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, QLD, 4006, Australia.,Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, 792 Elizabeth Street, Parkville, VIC, 3000, Australia
| | - Deborah Cromer
- Infection Analytics Program, Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, 2052, Australia
| | | | - R Kiplin Guy
- College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - James S McCarthy
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, QLD, 4006, Australia.,Department of Infectious Diseases, Peter Doherty Institute for Infection and Immunity, University of Melbourne, 792 Elizabeth Street, Parkville, VIC, 3000, Australia.,Victorian Infectious Diseases Service, Royal Melbourne Hospital, Parkville, VIC, 3000, Australia
| | - Miles P Davenport
- Infection Analytics Program, Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, 2052, Australia
| | - Ashraful Haque
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, Brisbane, QLD, 4006, Australia. .,Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, 792 Elizabeth Street, Parkville, VIC, 3000, Australia.
| | - David S Khoury
- Infection Analytics Program, Kirby Institute for Infection and Immunity, UNSW Sydney, Sydney, 2052, Australia.
| |
Collapse
|
9
|
Sun M, Xiao Q, Wang X, Yang C, Chen C, Tian X, Wang S, Li H, Qiu S, Shu J, Shou Y, Liang Y, Xue T, Chen K. Tumor-associated macrophages modulate angiogenesis and tumor growth in a xenograft mouse model of multiple myeloma. Leuk Res 2021; 110:106709. [PMID: 34560409 DOI: 10.1016/j.leukres.2021.106709] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/03/2021] [Accepted: 09/15/2021] [Indexed: 01/27/2023]
Abstract
Tumor-associated macrophages (TAMs) are closely associated with poor multiple myeloma (MM) prognosis. Therefore, in-depth understanding of the mechanism by which TAM supports MM progression may lead to its effective treatment. We used the MM nude mouse subcutaneous xenograft model to evaluate the efficacy of the macrophage-depleting agent clodronate liposome (Clo) against MM and elucidate the mode of action of this therapy. At the same time, observe whether the elimination of TAM in vivo while silencing the expression of VEGFA has the same effect as in vitro experiments. We also used Clo to eliminate macrophages and reinjected M1 or M2 TAM through mouse tail veins to investigate the effects of various macrophage subtypes on MM xenograft tumor growth. We applied qRT-PCR, immunohistochemistry, and enzyme-linked immunosorbent assay to quantify VEGFA, CD31, and CD163 expression in tumor tissues and sera. Removal of TAMs from the tumor microenvironment impeded tumor growth. The combination of Clo plus VEGFA siRNA had a stronger inhibitory effect on tumor growth than Clo alone, and M2 and M1 macrophages promoted and inhibited tumor growth, respectively. Macrophage depletion combined with cytokine blocking is a promising MM treatment. Targeted M2 macrophage elimination together with cytokine block may be more effective at inhibiting MM growth than either treatment alone. The results of the present study lay an empirical foundation for the development of novel therapeutic strategies for MM.
Collapse
Affiliation(s)
- Miaomiao Sun
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450052, Henan, People's Republic of China; Henan Province Key Laboratory of Tumor Pathology, Department of Pathology of The First Affiliated Hospital of Zhengzhou University, No. 40 Daxue Road, Zhengzhou, 450003, Henan, People's Republic of China
| | - Qiankun Xiao
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450052, Henan, People's Republic of China; School of Life Sciences, Zhengzhou University, No. 100 Kexue Da Dao, Zhengzhou, 450001, Henan, People's Republic of China; BGI College, Zhengzhou University, No. 40 Daxue Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Xiaoqian Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450052, Henan, People's Republic of China; BGI College, Zhengzhou University, No. 40 Daxue Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Chenbo Yang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Chao Chen
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Xiangyu Tian
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Shuaiyuan Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Hui Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Sen Qiu
- People's Hospital of Zhengzhou, Henan University of Chinese Medicine, No. 33 Huanghe Road, Zhengzhou, 410105, Henan, People's Republic of China
| | - Jiao Shu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450052, Henan, People's Republic of China; Henan Province Key Laboratory of Tumor Pathology, Department of Pathology of The First Affiliated Hospital of Zhengzhou University, No. 40 Daxue Road, Zhengzhou, 450003, Henan, People's Republic of China
| | - Yuwei Shou
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Yinghao Liang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Tong Xue
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450052, Henan, People's Republic of China
| | - Kuisheng Chen
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe Dong Road, Zhengzhou, 450052, Henan, People's Republic of China; Henan Province Key Laboratory of Tumor Pathology, Department of Pathology of The First Affiliated Hospital of Zhengzhou University, No. 40 Daxue Road, Zhengzhou, 450003, Henan, People's Republic of China.
| |
Collapse
|
10
|
Pack AD, Schwartzhoff PV, Zacharias ZR, Fernandez-Ruiz D, Heath WR, Gurung P, Legge KL, Janse CJ, Butler NS. Hemozoin-mediated inflammasome activation limits long-lived anti-malarial immunity. Cell Rep 2021; 36:109586. [PMID: 34433049 PMCID: PMC8432597 DOI: 10.1016/j.celrep.2021.109586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/21/2021] [Accepted: 07/30/2021] [Indexed: 12/04/2022] Open
Abstract
During acute malaria, most individuals mount robust inflammatory responses that limit parasite burden. However, long-lived sterilizing anti-malarial memory responses are not efficiently induced, even following repeated Plasmodium exposures. Using multiple Plasmodium species, genetically modified parasites, and combinations of host genetic and pharmacologic approaches, we find that the deposition of the malarial pigment hemozoin directly limits the abundance and capacity of conventional type 1 dendritic cells to prime helper T cell responses. Hemozoin-induced dendritic cell dysfunction results in aberrant Plasmodium-specific CD4 T follicular helper cell differentiation, which constrains memory B cell and long-lived plasma cell formation. Mechanistically, we identify that dendritic cell-intrinsic NLRP3 inflammasome activation reduces conventional type 1 dendritic cell abundance, phagocytosis, and T cell priming functions in vivo. These data identify biological consequences of hemozoin deposition during malaria and highlight the capacity of the malarial pigment to program immune evasion during the earliest events following an initial Plasmodium exposure.
Collapse
Affiliation(s)
- Angela D Pack
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | | | - Zeb R Zacharias
- Department of Pathology, University of Iowa, Iowa City, IA, USA
| | - Daniel Fernandez-Ruiz
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, VIC 3000, Australia
| | - William R Heath
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, VIC 3000, Australia; ARC Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, VIC 3010, Australia
| | - Prajwal Gurung
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
| | - Kevin L Legge
- Department of Pathology, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
| | - Chris J Janse
- Leiden Malaria Research Group, Centre of Infectious Diseases, Leiden University Medical Centre, Leiden 233 ZA, the Netherlands
| | - Noah S Butler
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
11
|
Djokic V, Rocha SC, Parveen N. Lessons Learned for Pathogenesis, Immunology, and Disease of Erythrocytic Parasites: Plasmodium and Babesia. Front Cell Infect Microbiol 2021; 11:685239. [PMID: 34414129 PMCID: PMC8369351 DOI: 10.3389/fcimb.2021.685239] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 07/15/2021] [Indexed: 11/24/2022] Open
Abstract
Malaria caused by Plasmodium species and transmitted by Anopheles mosquitoes affects large human populations, while Ixodes ticks transmit Babesia species and cause babesiosis. Babesiosis in animals has been known as an economic drain, and human disease has also emerged as a serious healthcare problem in the last 20–30 years. There is limited literature available regarding pathogenesis, immunity, and disease caused by Babesia spp. with their genomes sequenced only in the last decade. Therefore, using previous studies on Plasmodium as the foundation, we have compared similarities and differences in the pathogenesis of Babesia and host immune responses. Sexual life cycles of these two hemoparasites in their respective vectors are quite similar. An adult Anopheles female can take blood meal several times in its life such that it can both acquire and transmit Plasmodia to hosts. Since each tick stage takes blood meal only once, transstadial horizontal transmission from larva to nymph or nymph to adult is essential for the release of Babesia into the host. The initiation of the asexual cycle of these parasites is different because Plasmodium sporozoites need to infect hepatocytes before egressed merozoites can infect erythrocytes, while Babesia sporozoites are known to enter the erythrocytic cycle directly. Plasmodium metabolism, as determined by its two- to threefold larger genome than different Babesia, is more complex. Plasmodium replication occurs in parasitophorous vacuole (PV) within the host cells, and a relatively large number of merozoites are released from each infected RBC after schizogony. The Babesia erythrocytic cycle lacks both PV and schizogony. Cytoadherence that allows the sequestration of Plasmodia, primarily P. falciparum in different organs facilitated by prominent adhesins, has not been documented for Babesia yet. Inflammatory immune responses contribute to the severity of malaria and babesiosis. Antibodies appear to play only a minor role in the resolution of these diseases; however, cellular and innate immunity are critical for the clearance of both pathogens. Inflammatory immune responses affect the severity of both diseases. Macrophages facilitate the resolution of both infections and also offer cross-protection against related protozoa. Although the immunosuppression of adaptive immune responses by these parasites does not seem to affect their own clearance, it significantly exacerbates diseases caused by coinfecting bacteria during coinfections.
Collapse
Affiliation(s)
- Vitomir Djokic
- Department for Bacterial Zoonozes, Laboratory for Animal Health, French Agency for Food, Environmental and Occupational Health & Safety, UPEC, University Paris-Est, Maisons-Alfort, France
| | - Sandra C Rocha
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Nikhat Parveen
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, United States
| |
Collapse
|
12
|
Gnangnon B, Duraisingh MT, Buckee CO. Deconstructing the parasite multiplication rate of Plasmodium falciparum. Trends Parasitol 2021; 37:922-932. [PMID: 34119440 DOI: 10.1016/j.pt.2021.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 01/22/2023]
Abstract
Epidemiological indicators describing population-level malaria transmission dynamics are widely used to guide policy recommendations. However, the determinants of malaria outcomes within individuals are still poorly understood. This conceptual gap partly reflects the fact that there are few indicators that robustly predict the trajectory of individual infections or clinical outcomes. The parasite multiplication rate (PMR) is a widely used indicator for the Plasmodium intraerythrocytic development cycle (IDC), for example, but its relationship to clinical outcomes is complex. Here, we review its calculation and use in P. falciparum malaria research, as well as the parasite and host factors that impact it. We also provide examples of metrics that can help to link within-host dynamics to malaria clinical outcomes when used alongside the PMR.
Collapse
Affiliation(s)
- Bénédicte Gnangnon
- Center for Communicable Diseases Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Immunology & Infectious Diseases Department, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Manoj T Duraisingh
- Immunology & Infectious Diseases Department, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Caroline O Buckee
- Center for Communicable Diseases Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
13
|
Peters MAE, Greischar MA, Mideo N. Challenges in forming inferences from limited data: a case study of malaria parasite maturation. J R Soc Interface 2021; 18:20210065. [PMID: 33906391 DOI: 10.1098/rsif.2021.0065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Inferring biological processes from population dynamics is a common challenge in ecology, particularly when faced with incomplete data. This challenge extends to inferring parasite traits from within-host infection dynamics. We focus on rodent malaria infections (Plasmodium berghei), a system for which previous work inferred an immune-mediated extension in the length of the parasite development cycle within red blood cells. By developing a system of delay-differential equations to describe within-host infection dynamics and simulating data, we demonstrate the potential to obtain biased estimates of parasite (and host) traits when key biological processes are not considered. Despite generating infection dynamics using a fixed parasite developmental cycle length, we find that known sources of measurement bias in parasite stage and abundance data can affect estimates of parasite developmental duration, with stage misclassification driving inferences about extended cycle length. We discuss alternative protocols and statistical methods that can mitigate such misestimation.
Collapse
Affiliation(s)
- Madeline A E Peters
- Department of Ecology and Evolutionary Biology, The University of Toronto, Toronto Ontario, Canada
| | - Megan A Greischar
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY, USA
| | - Nicole Mideo
- Department of Ecology and Evolutionary Biology, The University of Toronto, Toronto Ontario, Canada
| |
Collapse
|
14
|
Yuguchi T, Kanoi BN, Nagaoka H, Miura T, Ito D, Takeda H, Tsuboi T, Takashima E, Otsuki H. Plasmodium yoelii Erythrocyte Binding Like Protein Interacts With Basigin, an Erythrocyte Surface Protein. Front Cell Infect Microbiol 2021; 11:656620. [PMID: 33937099 PMCID: PMC8079763 DOI: 10.3389/fcimb.2021.656620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/25/2021] [Indexed: 11/30/2022] Open
Abstract
Erythrocyte recognition and invasion is critical for the intra-erythrocytic development of Plasmodium spp. parasites. The multistep invasion process involves specific interactions between parasite ligands and erythrocyte receptors. Erythrocyte-binding-like (EBL) proteins, type I integral transmembrane proteins released from the merozoite micronemes, are known to play an important role in the initiation and formation of tight junctions between the apical end of the merozoite and the erythrocyte surface. In Plasmodium yoelii EBL (PyEBL), a single amino acid substitution in the putative Duffy binding domain dramatically changes parasite growth rate and virulence. This suggests that PyEBL is important for modulating the virulence of P. yoelii parasites. Based on these observations, we sought to elucidate the receptor of PyEBL that mediates its role as an invasion ligand. Using the eukaryotic wheat germ cell-free system, we systematically developed and screened a library of mouse erythrocyte proteins against native PyEBL using AlphaScreen technology. We report that PyEBL specifically interacts with basigin, an erythrocyte surface protein. We further confirmed that the N-terminal cysteine-rich Duffy binding-like region (EBL region 2), is responsible for the interaction, and that the binding is not affected by the C351Y mutation, which was previously shown to modulate virulence of P. yoelii. The identification of basigin as the putative PyEBL receptor offers new insights into the role of this molecule and provides an important base for in-depth studies towards developing novel interventions against malaria.
Collapse
Affiliation(s)
- Takaaki Yuguchi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Bernard N Kanoi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Toyokazu Miura
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Daisuke Ito
- Division of Medical Zoology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Hiroyuki Takeda
- Division of Proteo-Drug-Discovery Sciences, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Hitoshi Otsuki
- Division of Medical Zoology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
15
|
Rogers KJ, Vijay R, Butler NS. Anti-malarial humoral immunity: the long and short of it. Microbes Infect 2021; 23:104807. [PMID: 33684519 DOI: 10.1016/j.micinf.2021.104807] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/28/2021] [Accepted: 02/28/2021] [Indexed: 12/17/2022]
Abstract
Humoral immunity is critical for limiting Plasmodium parasite infections and the severity of malaria. Naturally acquired immunity against malaria occurs inefficiently and protection is relatively short-lived. Here we review recent advances and explore emerging hypotheses regarding the molecular and cellular pathways that regulate Plasmodium parasite-specific B cell responses and durable anti-malarial humoral immunity.
Collapse
Affiliation(s)
- Kai J Rogers
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Rahul Vijay
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA
| | - Noah S Butler
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Immunology, The University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
16
|
Furtado R, Chorro L, Zimmerman N, Guillen E, Spaulding E, Chin SS, Daily JP, Lauvau G. Blockade of LAG-3 in PD-L1-Deficient Mice Enhances Clearance of Blood Stage Malaria Independent of Humoral Responses. Front Immunol 2021; 11:576743. [PMID: 33519801 PMCID: PMC7840658 DOI: 10.3389/fimmu.2020.576743] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 12/03/2020] [Indexed: 11/23/2022] Open
Abstract
T cells expressing high levels of inhibitory receptors such as PD-1 and LAG-3 are a hallmark of chronic infections and cancer. Checkpoint blockade therapies targeting these receptors have been largely validated as promising strategies to restore exhausted T cell functions and clearance of chronic infections and tumors. The inability to develop long-term natural immunity in malaria-infected patients has been proposed to be at least partially accounted for by sustained expression of high levels of inhibitory receptors on T and B lymphocytes. While blockade or lack of PD-1/PD-L1 and/or LAG-3 was reported to promote better clearance of Plasmodium parasites in various mouse models, how exactly blockade of these pathways contributes to enhanced protection is not known. Herein, using the mouse model of non-lethal P. yoelii (Py) infection, we reveal that the kinetics of blood parasitemia as well as CD4+ T follicular helper (TFH) and germinal center (GC) B cell responses are indistinguishable between PD-1-/-, PD-L1-/- and WT mice. Yet, we also report that monoclonal antibody (mAb) blockade of LAG-3 in PD-L1-/- mice promotes accelerated control of blood parasite growth and clearance, consistent with prior therapeutic blockade experiments. However, neither CD4+ TFH and GC B cell responses, nor parasite-specific Ab serum titers and capacity to transfer protection differed. We also found that i) the majority of LAG-3+ cells are T cells, ii) selective depletion of CD4+ but not CD8+ T cells prevents anti-LAG-3-mediated protection, and iii) production of effector cytokines by CD4+ T cells is increased in anti-LAG-3-treated versus control mice. Thus, taken together, these results are consistent with a model in which blockade and/or deficiency of PD-L1 and LAG-3 on parasite-specific CD4+ T cells unleashes their ability to effectively clear blood parasites, independently from humoral responses.
Collapse
Affiliation(s)
- Raquel Furtado
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Laurent Chorro
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Natalie Zimmerman
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Erik Guillen
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Emily Spaulding
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Shu Shien Chin
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Johanna P Daily
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States.,Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Grégoire Lauvau
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
17
|
Transcriptome dynamics of CD4 + T cells during malaria maps gradual transit from effector to memory. Nat Immunol 2020; 21:1597-1610. [PMID: 33046889 DOI: 10.1038/s41590-020-0800-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 08/28/2020] [Indexed: 02/06/2023]
Abstract
The dynamics of CD4+ T cell memory development remain to be examined at genome scale. In malaria-endemic regions, antimalarial chemoprevention protects long after its cessation and associates with effects on CD4+ T cells. We applied single-cell RNA sequencing and computational modelling to track memory development during Plasmodium infection and treatment. In the absence of central memory precursors, two trajectories developed as T helper 1 (TH1) and follicular helper T (TFH) transcriptomes contracted and partially coalesced over three weeks. Progeny of single clones populated TH1 and TFH trajectories, and fate-mapping suggested that there was minimal lineage plasticity. Relationships between TFH and central memory were revealed, with antimalarials modulating these responses and boosting TH1 recall. Finally, single-cell epigenomics confirmed that heterogeneity among effectors was partially reset in memory. Thus, the effector-to-memory transition in CD4+ T cells is gradual during malaria and is modulated by antiparasitic drugs. Graphical user interfaces are presented for examining gene-expression dynamics and gene-gene correlations ( http://haquelab.mdhs.unimelb.edu.au/cd4_memory/ ).
Collapse
|
18
|
Stopping Plasmodium Parasite Invasion by Boosting Humoral Immunity. Hemasphere 2019; 3:e206. [PMID: 31723834 PMCID: PMC6746016 DOI: 10.1097/hs9.0000000000000206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
19
|
Kurtovic L, Boyle MJ, Opi DH, Kennedy AT, Tham WH, Reiling L, Chan JA, Beeson JG. Complement in malaria immunity and vaccines. Immunol Rev 2019; 293:38-56. [PMID: 31556468 PMCID: PMC6972673 DOI: 10.1111/imr.12802] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 09/03/2019] [Indexed: 12/20/2022]
Abstract
Developing efficacious vaccines for human malaria caused by Plasmodium falciparum is a major global health priority, although this has proven to be immensely challenging over the decades. One major hindrance is the incomplete understanding of specific immune responses that confer protection against disease and/or infection. While antibodies to play a crucial role in malaria immunity, the functional mechanisms of these antibodies remain unclear as most research has primarily focused on the direct inhibitory or neutralizing activity of antibodies. Recently, there is a growing body of evidence that antibodies can also mediate effector functions through activating the complement system against multiple developmental stages of the parasite life cycle. These antibody‐complement interactions can have detrimental consequences to parasite function and viability, and have been significantly associated with protection against clinical malaria in naturally acquired immunity, and emerging findings suggest these mechanisms could contribute to vaccine‐induced immunity. In order to develop highly efficacious vaccines, strategies are needed that prioritize the induction of antibodies with enhanced functional activity, including the ability to activate complement. Here we review the role of complement in acquired immunity to malaria, and provide insights into how this knowledge could be used to harness complement in malaria vaccine development.
Collapse
Affiliation(s)
- Liriye Kurtovic
- Burnet Institute, Melbourne, Vic., Australia.,Central Clinical School, Monash University, Melbourne, Vic., Australia
| | | | | | - Alexander T Kennedy
- Walter and Eliza Hall Institute, Melbourne, Vic., Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Vic., Australia
| | - Wai-Hong Tham
- Walter and Eliza Hall Institute, Melbourne, Vic., Australia
| | | | - Jo-Anne Chan
- Burnet Institute, Melbourne, Vic., Australia.,Central Clinical School, Monash University, Melbourne, Vic., Australia
| | - James G Beeson
- Burnet Institute, Melbourne, Vic., Australia.,Central Clinical School, Monash University, Melbourne, Vic., Australia.,Department of Microbiology, Monash University, Clayton, Vic., Australia.,Department of Medicine, The University of Melbourne, Parkville, Vic., Australia
| |
Collapse
|