1
|
Reiling L, Persson KEM, McCallum FJ, Gicheru N, Kinyanjui SM, Chitnis CE, Fowkes FJI, Marsh K, Beeson JG. Plasmodium falciparum reticulocyte-binding homologues are targets of human inhibitory antibodies and play a role in immune evasion. Front Immunol 2025; 16:1532451. [PMID: 40201183 PMCID: PMC11975925 DOI: 10.3389/fimmu.2025.1532451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/28/2025] [Indexed: 04/10/2025] Open
Abstract
Introduction Antibodies targeting the blood-stage of Plasmodium falciparum play a critical role in naturally acquired immunity to malaria by limiting blood-stage parasitemia. One mode of action of antibodies is the direct inhibition of merozoite invasion of erythrocytes through targeting invasion ligands. However, evasion of inhibitory antibodies may be mediated in P. falciparum by switching between various ligand-mediated merozoite invasion pathways. Here, we investigated the potential roles of invasion ligands PfRH1, PfRH2a and PfRH2b in immune evasion through phenotypic variation, and their importance as targets of human invasion-inhibitory antibodies. Methods Serum samples from malaria-exposed children and adults in Kenya were examined for their ability to inhibit P. falciparum invasion, using parasites with disrupted pfrh1, pfrh2a or pfrh2b genes. Results and Discussion The loss of PfRH1 and PfRH2b substantially impacted on susceptibility to inhibitory antibodies, suggesting that variation in the use of these ligands contributes to immune evasion. The effect was less prominent with loss of PfRH2a. Differential inhibition of the knockout and parental lines points to PfRH1 and PfRH2b as targets of acquired growth inhibitory antibodies whereas PfRH2a appeared to be a minor target. There was limited relatedness of the inhibitory responses between different isolates or compared to parasites with deletions of erythrocyte-binding antigens. This further suggests that there is a substantial amount of antigenic diversity in invasion pathways to facilitate immune evasion. These findings provide evidence that PfRH1 and PfRH2b are significant targets of inhibitory antibodies and variation in their expression may facilitate immune evasion. Targeting of multiple invasion ligands in vaccine design is likely to be required to achieve potent inhibitory antibodies and protective efficacy against malaria.
Collapse
Affiliation(s)
- Linda Reiling
- Department of Life Sciences, Burnet Institute of Medical Research and Public Health, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, VIC, Australia
- Department of Immunology, Monash University, Melbourne, VIC, Australia
| | - Kristina E. M. Persson
- Department of Laboratory Medicine, Lund University, Lund, Sweden
- Clinical Chemistry and Pharmacology, Skåne University Hospital, Lund, Sweden
| | - Fiona J. McCallum
- Australian Defence Force Malaria and Infectious Disease Institute, Enoggera, QLD, Australia
| | - Nimmo Gicheru
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute - Wellcome Trust Research Programme, Kilifi, Kenya
| | - Samson M. Kinyanjui
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute - Wellcome Trust Research Programme, Kilifi, Kenya
| | - Chetan E. Chitnis
- Department of Parasites and Insect Vectors, Pasteur Institute, Paris, France
| | - Freya J. I. Fowkes
- Department of Life Sciences, Burnet Institute of Medical Research and Public Health, Melbourne, VIC, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Kevin Marsh
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute - Wellcome Trust Research Programme, Kilifi, Kenya
- Nuffield Department of Medicine, Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Oxford, United Kingdom
| | - James G. Beeson
- Department of Life Sciences, Burnet Institute of Medical Research and Public Health, Melbourne, VIC, Australia
- Department of Microbiology, Monash University, Melbourne, VIC, Australia
- School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Department of Infectious Diseases, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
2
|
Odhiambo EO, Mellencamp KA, Ondigo BN, Hamre KES, Beeson JG, Opi DH, Narum DL, Ayodo G, John CC. Antibody correlates of risk of clinical malaria in an area of low and unstable malaria transmission in western Kenya. Malar J 2025; 24:73. [PMID: 40033373 PMCID: PMC11877692 DOI: 10.1186/s12936-025-05300-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/18/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Defining antibody correlates of protection against clinical malaria in areas of low and unstable transmission is challenging because of limited malaria cases in these areas. Additionally, clinical malaria affects both adults and children in areas of low and unstable transmission, but it is unclear whether antibody correlates of protection against malaria differ with age. METHODS Blood samples were obtained from 5753 individuals in Kenyan highland area with low and seasonal malaria transmission in 2007 and recorded episodes of clinical malaria in this population from 2007 to 2017. Using a nested case-control study design, participants who developed clinical malaria (cases) were matched by age and village to those who did not (controls). Immunoglobulin (Ig)G, IgG1, IgG3, IgA and IgM responses to 16 Plasmodium falciparum antigens were compared in individuals < 5 years old (80 cases vs. 240 controls), 5-14 years old (103 cases vs. 309 controls) and ≥ 15 years old (118 cases vs. 354 controls). Antibody level was correlated with risk of clinical malaria, adjusted for malaria exposure markers. RESULTS In all age groups, most antibodies were not associated with risk of clinical malaria. In children < 5 years, higher levels of IgG to GLURP-R2 and MSP-2, IgG1 to GLURP-R2, and IgG3 to MSP-2 were associated with reduced risk of clinical malaria, while higher IgG3 levels to CSP were associated with increased risk of clinical malaria. In children 5-14 years and individuals ≥ 15 years, higher antibody levels to multiple P. falciparum antigens were associated with an increased risk of clinical malaria, and none were associated with decreased risk of clinical malaria. CONCLUSIONS Antibody correlates of protection against clinical malaria were observed only in children < 5 years old in this area of low and unstable malaria transmission. In older children and adults in this area, some antibody responses correlated with increased risk of clinical malaria. Future studies in low malaria transmission areas should evaluate the comparative contributions of cellular and humoral immunity to protection from clinical malaria in young children versus older children and adults.
Collapse
Affiliation(s)
- Eliud O Odhiambo
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, USA
| | - Kagan A Mellencamp
- Ryan White Center for Pediatric Infectious Diseases & Global Health, Indiana University School of Medicine, Indianapolis, USA
| | - Bartholomew N Ondigo
- Department of Biochemistry and Molecular Biology, Egerton University, Nakuru, Kenya
| | | | | | | | - David L Narum
- National Institutes of Health (NIAID/NIH), National Institute of Allergy and Infectious Diseases, Maryland, USA
| | - George Ayodo
- Jaramogi Oginga Odinga University of Science and Technology, Bondo, Kenya
| | - Chandy C John
- Ryan White Center for Pediatric Infectious Diseases & Global Health, Indiana University School of Medicine, Indianapolis, USA.
| |
Collapse
|
3
|
Soares IF, de Oliveira Baptista B, da Silva Matos A, Rodrigues-da-Silva RN, Kujbida Junior MA, Albrecht L, Rodolphi CM, Scopel KKG, Alencar ALC, de Souza RM, Dos Santos de Souza HA, Riccio EKP, de Barros JP, Totino PRR, Daniel-Ribeiro CT, Pratt-Riccio LR, Lima-Junior JDC. Characterization of T and B cell epitopes in PvCyRPA by studying the naturally acquired immune response in Brazilian Amazon communities. Sci Rep 2024; 14:27343. [PMID: 39521783 PMCID: PMC11550457 DOI: 10.1038/s41598-024-72671-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/09/2024] [Indexed: 11/16/2024] Open
Abstract
Plasmodium vivax, a challenging species to eliminate, causes millions of malaria cases globally annually. Developing an effective vaccine is crucial in the fight against vivax malaria, but considering the limited number of studies focusing on the identification and development of P. vivax-specific vaccine candidates, exploring new antigens is an urgent need. The merozoite protein CyRPA is essential for P. falciparum growth and erythrocyte invasion and corresponds to a promising candidate antigen. In P. vivax, a single study with multiple vaccine candidates indicates PvCyRPA with strong association with protection, outperforming classic malaria vaccine candidates. However, little is known about the specific naturally acquired response in the Americas, as well as the antigen epitope mapping. For this reason, we aimed to investigate the cellular and humoral immune response elicited against PvCyRPA in Brazilian endemic areas to identify the existence of immunodominant regions and the potential of this protein as a single or even a multi-stage specific malaria vaccine candidate for P. vivax. The results demonstrated that PvCyRPA is naturally immunogenic in Brazilian Amazon individuals previously exposed to malaria, which presented anti-PvCyRPA cytophilic antibodies. Moreover, our data show that the protein also possesses important immunogenic regions with an overlap of B and T cell epitopes. These data reinforce the possibility of including PvCyRPA in vaccine formulations for P. vivax.
Collapse
Affiliation(s)
- Isabela Ferreira Soares
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz, (Fiocruz), Rio de Janeiro, RJ, Brazil
| | | | - Ada da Silva Matos
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz, (Fiocruz), Rio de Janeiro, RJ, Brazil
| | | | | | - Letusa Albrecht
- Laboratório de Pesquisa em Apicomplexa, Instituto Carlos Chagas, Fiocruz, Curitiba, PR, Brazil
| | | | | | - Ana Luiza Carneiro Alencar
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz, (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Rodrigo Medeiros de Souza
- Laboratório de Doenças infecciosas na Amazônia Ocidental - Universidade Federal do Acre, Cruzeiro do Sul, AC, Brazil
| | | | | | | | | | - Cláudio Tadeu Daniel-Ribeiro
- Laboratório de Pesquisa em Malária, IOC, Fiocruz, Rio de Janeiro, RJ, Brazil
- Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal), Secretaria de Vigilância em Saúde e Ambiente (SVSA), Ministério da Saúde, Rio de Janeiro, RJ, Brazil
| | | | - Josué da Costa Lima-Junior
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz, (Fiocruz), Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
4
|
Tukwasibwe S, Lewis SN, Taremwa Y, van der Ploeg K, Press KD, Ty M, Namirimu Nankya F, Musinguzi K, Nansubuga E, Bach F, Chamai M, Okitwi M, Tumusiime G, Nakimuli A, Colucci F, Kamya MR, Nankabirwa JI, Arinaitwe E, Greenhouse B, Dorsey G, Rosenthal PJ, Ssewanyana I, Jagannathan P. Natural killer cell antibody-dependent cellular cytotoxicity to Plasmodium falciparum is impacted by cellular phenotypes, erythrocyte polymorphisms, parasite diversity and intensity of transmission. Clin Transl Immunology 2024; 13:e70005. [PMID: 39493859 PMCID: PMC11528551 DOI: 10.1002/cti2.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 08/09/2024] [Accepted: 09/12/2024] [Indexed: 11/05/2024] Open
Abstract
Objectives Natural killer (NK) cells make important contributions to anti-malarial immunity through antibody-dependent cellular cytotoxicity (ADCC), but the role of different components of this pathway in promoting NK cell activation remains unclear. Methods We compared the functions and phenotypes of NK cells from malaria-exposed and malaria-naive donors, and then varied the erythrocyte genetic background, Plasmodium falciparum strain and opsonising plasma used in ADCC to observe their impacts on NK cell degranulation as measured by CD107a mobilisation. Results Natural killer cells from malaria-exposed adult Ugandan donors had enhanced ADCC, but an impaired pro-inflammatory response to cytokine stimulation, compared to NK cells obtained from malaria-naive adult North American donors. Cellular phenotypes from malaria-exposed donors reflected this specialisation for ADCC, with a compartment-wide downregulation of the Fc receptor γ-chain and enrichment of highly differentiated CD56dim and CD56neg populations. NK cell degranulation was enhanced in response to opsonised P. falciparum schizonts cultured in sickle cell heterozygous erythrocytes relative to wild-type erythrocytes, and when using opsonising plasma collected from donors living in a high transmission area compared to a lower transmission area despite similar levels of 3D7 schizont-specific IgG levels. However, degranulation was lowered in response to opsonised field isolate P. falciparum schizonts isolated from clinical malaria infections, compared to the 3D7 laboratory strain typically used in these assays. Conclusion This work highlights important host and parasite factors that contribute to ADCC efficacy that should be considered in the design of ADCC assays.
Collapse
Affiliation(s)
- Stephen Tukwasibwe
- Infectious Diseases Research CollaborationKampalaUganda
- School of Medicine, Uganda Christian UniversityMukonoUganda
| | | | | | | | | | - Maureen Ty
- Department of MedicineStanford UniversityStanfordCAUSA
| | | | | | | | - Florian Bach
- Department of MedicineStanford UniversityStanfordCAUSA
| | - Martin Chamai
- Infectious Diseases Research CollaborationKampalaUganda
| | - Martin Okitwi
- Infectious Diseases Research CollaborationKampalaUganda
| | | | | | - Francesco Colucci
- Department of Obstetrics and GynaecologyUniversity of CambridgeCambridgeUK
| | - Moses R Kamya
- Infectious Diseases Research CollaborationKampalaUganda
- School of Medicine, Makerere UniversityKampalaUganda
| | - Joaniter I Nankabirwa
- Infectious Diseases Research CollaborationKampalaUganda
- School of Medicine, Makerere UniversityKampalaUganda
| | | | - Bryan Greenhouse
- Department of MedicineUniversity of California San FranciscoSan FranciscoCAUSA
| | - Grant Dorsey
- Department of MedicineUniversity of California San FranciscoSan FranciscoCAUSA
| | - Philip J Rosenthal
- Department of MedicineUniversity of California San FranciscoSan FranciscoCAUSA
| | | | | |
Collapse
|
5
|
Rathay V, Fürle K, Kiehl V, Ulmer A, Lanzer M, Thomson-Luque R. IgG Subclass Switch in Volunteers Repeatedly Immunized with the Full-Length Plasmodium falciparum Merozoite Surface Protein 1 (MSP1). Vaccines (Basel) 2024; 12:208. [PMID: 38400191 PMCID: PMC10893298 DOI: 10.3390/vaccines12020208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Vaccines are highly effective tools against infectious diseases and are also considered necessary in the fight against malaria. Vaccine-induced immunity is frequently mediated by antibodies. We have recently conducted a first-in-human clinical trial featuring SumayaVac-1, a malaria vaccine based on the recombinant, full-length merozoite surface protein 1 (MSP1FL) formulated with GLA-SE as an adjuvant. Vaccination with MSP1FL was safe and elicited sustainable IgG antibody titers that exceeded those observed in semi-immune populations from Africa. Moreover, IgG antibodies stimulated various Fc-mediated effector mechanisms associated with protection against malaria. However, these functionalities gradually waned. Here, we show that the initial two doses of SumayaVac-1 primarily induced the cytophilic subclasses IgG1 and IgG3. Unexpectedly, a shift in the IgG subclass composition occurred following the third and fourth vaccinations. Specifically, there was a progressive transition to IgG4 antibodies, which displayed a reduced capacity to engage in Fc-mediated effector functions and also exhibited increased avidity. In summary, our analysis of antibody responses to MSP1FL vaccination unveils a temporal shift towards noninflammatory IgG4 antibodies. These findings underscore the importance of considering the impact of IgG subclass composition on vaccine-induced immunity, particularly concerning Fc-mediated effector functions. This knowledge is pivotal in guiding the design of optimal vaccination strategies against malaria, informing decision making for future endeavors in this critical field.
Collapse
Affiliation(s)
- Veronika Rathay
- Parasitology, Centre for Infectious Diseases, University Hospital Heidelberg, Medical Faculty, Heidelberg University, 69120 Heidelberg, Germany
| | - Kristin Fürle
- Parasitology, Centre for Infectious Diseases, University Hospital Heidelberg, Medical Faculty, Heidelberg University, 69120 Heidelberg, Germany
| | - Viktoria Kiehl
- Parasitology, Centre for Infectious Diseases, University Hospital Heidelberg, Medical Faculty, Heidelberg University, 69120 Heidelberg, Germany
| | - Anne Ulmer
- Parasitology, Centre for Infectious Diseases, University Hospital Heidelberg, Medical Faculty, Heidelberg University, 69120 Heidelberg, Germany
| | - Michael Lanzer
- Parasitology, Centre for Infectious Diseases, University Hospital Heidelberg, Medical Faculty, Heidelberg University, 69120 Heidelberg, Germany
| | - Richard Thomson-Luque
- Parasitology, Centre for Infectious Diseases, University Hospital Heidelberg, Medical Faculty, Heidelberg University, 69120 Heidelberg, Germany
- Sumaya-Biotech GmbH & Co. KG, 69115 Heidelberg, Germany
| |
Collapse
|
6
|
Thawornpan P, Nicholas J, Malee C, Kochayoo P, Wangriatisak K, Tianpothong P, Ntumngia FB, J. Barnes S, H. Adams J, Chootong P. Longitudinal analysis of antibody responses to Plasmodium vivax sporozoite antigens following natural infection. PLoS Negl Trop Dis 2024; 18:e0011907. [PMID: 38277340 PMCID: PMC10817200 DOI: 10.1371/journal.pntd.0011907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 01/08/2024] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND P. vivax malaria is a major global health burden hindering social and economic development throughout many tropical and sub-tropical countries. Pre-erythrocytic (PE) vaccines emerge as an attractive approach for the control and elimination of malaria infection. Therefore, evaluating the magnitude, longevity and prevalence of naturally acquired IgG antibody responses against PE candidate antigens is useful for vaccine design. METHODOLOGY/PRINCIPAL FINDINGS The antigenicity of five recombinant PE antigens (PvCSP-VK210, PvSSP3, PvM2-MAEBL, PvCelTOS and PvSPECT1) was evaluated in plasma samples from individuals residing in low transmission areas in Thailand (Ranong and Chumphon Provinces). The samples were collected at the time of acute vivax malaria and 90, 270 and 360 days later. The prevalence, magnitude and longevity of total IgG and IgG subclasses were determined for each antigen using the longitudinal data. Our results showed that seropositivity of all tested PE antigens was detected during infection in at least some subjects; anti-PvCSP-VK210 and anti-PvCelTOS antibodies were the most frequent. Titers of these antibodies declined during the year of follow up, but notably seropositivity persisted. Among seropositive subjects at post-infection, high number of subjects possessed antibodies against PvCSP-VK210. Anti-PvSSP3 antibody responses had the longest half-life. IgG subclass profiling showed that the predominant subclasses were IgG1 and IgG3 (cytophilic antibodies), tending to remain detectable for at least 360 days after infection. CONCLUSIONS/SIGNIFICANCE The present study demonstrated the magnitude and longevity of serological responses to multiple PE antigens of P. vivax after natural infection. This knowledge could contribute to the design of an effective P. vivax vaccine.
Collapse
Affiliation(s)
- Pongsakorn Thawornpan
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Justin Nicholas
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Chayapat Malee
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Piyawan Kochayoo
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Kittikorn Wangriatisak
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Pachara Tianpothong
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Francis Babila Ntumngia
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Samantha J. Barnes
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - John H. Adams
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Patchanee Chootong
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| |
Collapse
|
7
|
Walker IS, Rogerson SJ. Pathogenicity and virulence of malaria: Sticky problems and tricky solutions. Virulence 2023; 14:2150456. [PMID: 36419237 PMCID: PMC9815252 DOI: 10.1080/21505594.2022.2150456] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/25/2022] Open
Abstract
Infections with Plasmodium falciparum and Plasmodium vivax cause over 600,000 deaths each year, concentrated in Africa and in young children, but much of the world's population remain at risk of infection. In this article, we review the latest developments in the immunogenicity and pathogenesis of malaria, with a particular focus on P. falciparum, the leading malaria killer. Pathogenic factors include parasite-derived toxins and variant surface antigens on infected erythrocytes that mediate sequestration in the deep vasculature. Host response to parasite toxins and to variant antigens is an important determinant of disease severity. Understanding how parasites sequester, and how antibody to variant antigens could prevent sequestration, may lead to new approaches to treat and prevent disease. Difficulties in malaria diagnosis, drug resistance, and specific challenges of treating P. vivax pose challenges to malaria elimination, but vaccines and other preventive strategies may offer improved disease control.
Collapse
Affiliation(s)
- Isobel S Walker
- Department of Infectious Diseases, The University of Melbourne, The Doherty Institute, Melbourne, Australia
| | - Stephen J Rogerson
- Department of Infectious Diseases, The University of Melbourne, The Doherty Institute, Melbourne, Australia
| |
Collapse
|
8
|
Fall AKDJ, Dechavanne C, Sabbagh A, Garcia A, Courtin D, Migot-Nabias F. Combined polymorphisms involving the IgG heavy chain and fc gamma receptors among Fulani and non-Fulani in Benin: Implications for the natural protection of young Fulani against plasmodium. Falciparum malaria infections. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2023:105461. [PMID: 37269963 DOI: 10.1016/j.meegid.2023.105461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/28/2023] [Accepted: 05/29/2023] [Indexed: 06/05/2023]
Abstract
A decreased susceptibility of Fulani populations to malaria infections has been shown in Africa. A previous longitudinal cohort study conducted in the Atacora region of northern Benin showed a high merozoite-phagocytosis capacity in young Fulani. Here, we explored the combined polymorphisms in the constant region of the IgG3 heavy chain (presence/absence of the G3m6 allotype) and in Fc gamma receptors (FcγRs) as potentially involved in the natural protection against malaria of young Fulani in Benin. An active malaria follow-up was conducted among individuals from Fulani, Bariba, Otamari and Gando ethnic groups living in sympatry in Atacora, over the full malaria transmission season. FcγRIIA 131R/H (rs1801274), FcγRIIC C/T (rs3933769) and FcγRIIIA 176F/V (rs396991) were determined using the TaqMan method; FcγRIIIB NA1/NA2 was assessed by polymerase chain reaction (PCR) using allele-specific primers and G3m6 using allotype by PCR-RFLP. Individual carriage of G3m6 (+) was associated with an increased risk of Pf malaria infection (logistic multivariate regression model (lmrm), IRR = 2.25, 95% CI = 1.06;4.74, P = 0.034). Combined haplotype G3m6 (+) - FcγRIIA 131H - FcγRIIC T - FcγRIIIA 176F - FcγRIIIB NA2 was also associated with an increased risk of Pf malaria infection (lmrm, IRR = 13.01, 95% CI = 1.69;99.76, P = 0.014). G3m6 (-), FcγRIIA 131R and FcγRIIIB NA1 were more prevalent in young Fulani (P = 0.002, P < 0.001 and P = 0.049, respectively), while no Fulani presented the combined G3m6 (+) - FcγRIIA 131H - FcγRIIC T - FcγRIIIA 176F - FcγRIIIB NA2 haplotype that was carried by a majority of infected children. Our results highlight the combined factors G3m6 - FcγR as potentially involved in the merozoite-phagocytosis capacity and in the natural protection of young Fulani individuals against P. falciparum malaria in Benin.
Collapse
Affiliation(s)
| | - Célia Dechavanne
- Université Paris Cité, IRD, UMR261, MERIT, F-75006 Paris, France
| | - Audrey Sabbagh
- Université Paris Cité, IRD, UMR261, MERIT, F-75006 Paris, France
| | - André Garcia
- Université Paris Cité, IRD, UMR261, MERIT, F-75006 Paris, France
| | - David Courtin
- Université Paris Cité, IRD, UMR261, MERIT, F-75006 Paris, France
| | | |
Collapse
|
9
|
Susceptibility to malaria in fulani, Bariba, Otamari and gando individuals living in sympatry in Benin: Role of opsonizing antibodies to Plasmodium falciparum merozoites. Heliyon 2023; 9:e13092. [PMID: 36711279 PMCID: PMC9879790 DOI: 10.1016/j.heliyon.2023.e13092] [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: 08/01/2022] [Revised: 12/17/2022] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
Objectives Fulani in Africa are known to be less susceptible to Plasmodium falciparum (Pf) malaria. This study explored a potential involvement of antibody-mediated merozoite phagocytosis mechanism in this natural protection against malaria. Methods Before the start of the malaria transmission season (MTS) in Benin, the functionality of antibodies against Pf merozoites was determined by the opsonic phagocytosis (OP) assay in plasma samples from Fulani, Bariba, Otamari and Gando groups. These individuals were actively followed-up for malaria detection from the beginning to the end of MTS. Anti-GLURP Immunoglobulin G antibody quantification, malaria Rapid Diagnostic Test (RDT) and spleen palpation were performed before and after MTS. Results In Bariba, Otamari and Gando, but not in Fulani, plasma from adults promoted higher levels of OP than the children (P = 0.003; P = 0.012; P = 0.031 and P = 0.122). A high proportion of Fulani children had higher OP and anti-GLURP (P < 0.0001) antibody levels as compared to non-Fulani children; whereas this was not observed for Fulani adults (P = 0.223). High OP levels before MTS were significantly related to negative RDT after MTS (P = 0.011). Conclusion Our results highlight the ability of opsonizing antibodies to potentially enhance natural protection of young Fulani individuals against Pf malaria in Benin.
Collapse
|
10
|
Fall AKDJ, Kana IH, Dechavanne C, Garcia-Senosiain A, Guitard E, Milet J, Massougbodji A, Garcia A, Dugoujon JM, Migot-Nabias F, Theisen M, Courtin D. Naturally acquired antibodies from Beninese infants promote Plasmodium falciparum merozoite-phagocytosis by human blood leukocytes: implications for control of asymptomatic malaria infections. Malar J 2022; 21:356. [PMCID: PMC9707106 DOI: 10.1186/s12936-022-04361-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 11/03/2022] [Indexed: 11/30/2022] Open
Abstract
Abstract
Background
Immunoglobulin G (IgG) antibodies are thought to play important roles in the protection against Plasmodium falciparum (P. falciparum) malaria. A longitudinal cohort study performed in the Southern part of Benin, identified a group of infants who were able to control asymptomatic malaria infections (CAIG).
Methods
IgG antibodies against distinct merozoite antigens were quantified in plasma from Beninese infants. Functionality of these antibodies was assessed by the merozoite-phagocytosis assay using THP-1 cells and primary neutrophils as effector cells. Gm allotypes were determined by a serological method of haemagglutination inhibition.
Results
Purified IgG from infants in CAIG promoted higher levels of merozoite-phagocytosis than did IgG from children who were unable to control asymptomatic infections (Ologit multivariate regression model, Coef. = 0.06, 95% CI 0.02;0.10, P = 0.002). High level of merozoite-phagocytosis activity was significantly associated with high levels of IgG against AMA1 (Coef. = 1.76, 95% CI 0.39;3.14, P = 0.012) and GLURP-R2 (Coef. = 12.24, 95% CI 1.35;23.12, P = 0.028). Moreover, infants of the G3m5,6,10,11,13,14,24 phenotype showed higher merozoite-phagocytosis activity (Generalized linear model multivariate regression, Coef. = 7.46, 95% CI 0.31;14.61, P = 0.041) than those presenting other G3m phenotypes.
Conclusion
The results of the present study confirm the importance of antibodies to merozoite surface antigens in the control of asymptomatic malaria infection in Beninese infants. The study also demonstrated that G3m phenotypes impact the functional activity of IgG. This last point could have a considerable impact in the research of candidate vaccines against malaria parasites or other pathogens.
Collapse
|
11
|
Buendía-González FO, Legorreta-Herrera M. The Similarities and Differences between the Effects of Testosterone and DHEA on the Innate and Adaptive Immune Response. Biomolecules 2022; 12:biom12121768. [PMID: 36551196 PMCID: PMC9775255 DOI: 10.3390/biom12121768] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/08/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
Androgens are steroids that modulate various processes in the body, ranging from reproduction, metabolism, and even immune response. The main androgens are testosterone, dihydrotestosterone (DHT) and dehydroepiandrosterone (DHEA). These steroids modulate the development and function of immune response cells. Androgens are generally attributed to immunosuppressive effects; however, this is not always the case. Variations in the concentrations of these hormones induce differences in the innate, humoral, and cell-mediated immune response, which is concentration dependent. The androgens at the highest concentration in the organism that bind to the androgen receptor (AR) are DHEA and testosterone. Therefore, in this work, we review the effects of DHEA and testosterone on the immune response. The main findings of this review are that DHEA and testosterone induce similar but also opposite effects on the immune response. Both steroids promote the activation of regulatory T cells, which suppresses the Th17-type response. However, while testosterone suppresses the inflammatory response, DHEA promotes it, and this modulation is important for understanding the involvement of androgens in infectious (bacterial, viral and parasitic) and autoimmune diseases, as well as in the sexual dimorphism that occurs in these diseases.
Collapse
Affiliation(s)
- Fidel Orlando Buendía-González
- Laboratorio de Inmunología Molecular, Unidad de Investigación Química Computacional, Síntesis y Farmacología de Moléculas de Interés Biológico, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Iztapalapa, Ciudad de México 09230, Mexico
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Edificio D, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, Ciudad de México 04510, Mexico
| | - Martha Legorreta-Herrera
- Laboratorio de Inmunología Molecular, Unidad de Investigación Química Computacional, Síntesis y Farmacología de Moléculas de Interés Biológico, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Iztapalapa, Ciudad de México 09230, Mexico
- Correspondence:
| |
Collapse
|
12
|
Liu ZSJ, Sattabongkot J, White M, Chotirat S, Kumpitak C, Takashima E, Harbers M, Tham WH, Healer J, Chitnis CE, Tsuboi T, Mueller I, Longley RJ. Naturally acquired antibody kinetics against Plasmodium vivax antigens in people from a low malaria transmission region in western Thailand. BMC Med 2022; 20:89. [PMID: 35260169 PMCID: PMC8904165 DOI: 10.1186/s12916-022-02281-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 02/02/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Plasmodium vivax (P. vivax) is the dominant Plasmodium spp. causing the disease malaria in low-transmission regions outside of Africa. These regions often feature high proportions of asymptomatic patients with sub-microscopic parasitaemia and relapses. Naturally acquired antibody responses are induced after Plasmodium infection, providing partial protection against high parasitaemia and clinical episodes. However, previous work has failed to address the presence and maintenance of such antibody responses to P. vivax particularly in low-transmission regions. METHODS We followed 34 patients in western Thailand after symptomatic P. vivax infections to monitor antibody kinetics over 9 months, during which no recurrent infections occurred. We assessed total IgG, IgG subclass and IgM levels to up to 52 P. vivax proteins every 2-4 weeks using a multiplexed Luminex® assay and identified protein-specific variation in antibody longevity. Mathematical modelling was used to generate the estimated half-life of antibodies, long-, and short-lived antibody-secreting cells. RESULTS Generally, an increase in antibody level was observed within 1-week post symptomatic infection, followed by an exponential decay of different rates. We observed mostly IgG1 dominance and IgG3 sub-dominance in this population. IgM responses followed similar kinetic patterns to IgG, with some proteins unexpectedly inducing long-lived IgM responses. We also monitored antibody responses against 27 IgG-immunogenic antigens in 30 asymptomatic individuals from a similar region. Our results demonstrate that most antigens induced robust and long-lived total IgG responses following asymptomatic infections in the absence of (detected) boosting infections. CONCLUSIONS Our work provides new insights into the development and maintenance of naturally acquired immunity to P. vivax and will guide the potential use of serology to indicate immune status and/or identify populations at risk.
Collapse
Affiliation(s)
- Zoe Shih-Jung Liu
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, 3010, Australia
- Current affiliation: Deakin University, School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Geelong, Victoria, 3220, Australia
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Michael White
- Infectious Disease Epidemiology and Analytics G5 Unit, Department of Global Health, Institut Pasteur, Paris, France
| | - Sadudee Chotirat
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Chalermpon Kumpitak
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Eizo Takashima
- Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Matthias Harbers
- CellFree Sciences Co., Ltd., Yokohama, Japan and RIKEN Centre for Integrative Medical Sciences, Yokohama, Japan
| | - Wai-Hong Tham
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Julie Healer
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Chetan E Chitnis
- Malaria Parasite Biology and Vaccines, Department of Parasites & Insect Vectors, Institut Pasteur, Paris, France
| | | | - Ivo Mueller
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Rhea J Longley
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
13
|
Walker IS, Chung AW, Damelang T, Rogerson SJ. Analysis of Antibody Reactivity to Malaria Antigens by Microsphere-Based Multiplex Immunoassay. Methods Mol Biol 2022; 2470:309-325. [PMID: 35881355 DOI: 10.1007/978-1-0716-2189-9_23] [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] [Indexed: 06/15/2023]
Abstract
Protein multiplex assays enable serological analysis of multiple target proteins simultaneously, using relatively small volumes of patient sample per assay. Here we present a detailed protocol to analyze antibody reactivity to malaria antigens by microsphere-based multiplex assay (xMAP technology). This method involves coupling of recombinant proteins to fluorescently labeled microspheres; simultaneous exposure of all microspheres to plasma or sera, and detection of antigen-specific antibodies with a fluorescent labeled anti-human Fc region antibody. In addition to total IgG, this assay can be adapted to measure multiple properties of the antibody Fc region, including subclass, isotype, and Fc receptor or complement C1q binding.
Collapse
Affiliation(s)
- Isobel S Walker
- Department of Medicine, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
| | - Amy W Chung
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Timon Damelang
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Stephen J Rogerson
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
| |
Collapse
|
14
|
Bonam SR, Rénia L, Tadepalli G, Bayry J, Kumar HMS. Plasmodium falciparum Malaria Vaccines and Vaccine Adjuvants. Vaccines (Basel) 2021; 9:1072. [PMID: 34696180 PMCID: PMC8541031 DOI: 10.3390/vaccines9101072] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/10/2021] [Accepted: 09/22/2021] [Indexed: 12/02/2022] Open
Abstract
Malaria-a parasite vector-borne disease-is a global health problem, and Plasmodium falciparum has proven to be the deadliest among Plasmodium spp., which causes malaria in humans. Symptoms of the disease range from mild fever and shivering to hemolytic anemia and neurological dysfunctions. The spread of drug resistance and the absence of effective vaccines has made malaria disease an ever-emerging problem. Although progress has been made in understanding the host response to the parasite, various aspects of its biology in its mammalian host are still unclear. In this context, there is a pressing demand for the development of effective preventive and therapeutic strategies, including new drugs and novel adjuvanted vaccines that elicit protective immunity. The present article provides an overview of the current knowledge of anti-malarial immunity against P. falciparum and different options of vaccine candidates in development. A special emphasis has been made on the mechanism of action of clinically used vaccine adjuvants.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Equipe-Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, Université de Paris, F-75006 Paris, France;
| | - Laurent Rénia
- A*STAR Infectious Diseases Labs, 8A Biomedical Grove, Singapore 138648, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore 308232, Singapore
| | - Ganesh Tadepalli
- Vaccine Immunology Laboratory, Organic Synthesis and Process Chemistry Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India;
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Equipe-Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, Université de Paris, F-75006 Paris, France;
- Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad 678623, India
| | - Halmuthur Mahabalarao Sampath Kumar
- Vaccine Immunology Laboratory, Organic Synthesis and Process Chemistry Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India;
| |
Collapse
|
15
|
Willcox AC, Huber AS, Diouf A, Barrett JR, Silk SE, Pulido D, King LDW, Alanine DGW, Minassian AM, Diakite M, Draper SJ, Long CA, Miura K. Antibodies from malaria-exposed Malians generally interact additively or synergistically with human vaccine-induced RH5 antibodies. CELL REPORTS MEDICINE 2021; 2:100326. [PMID: 34337556 PMCID: PMC8324462 DOI: 10.1016/j.xcrm.2021.100326] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/03/2021] [Accepted: 06/04/2021] [Indexed: 11/26/2022]
Abstract
Reticulocyte-binding protein homolog 5 (RH5) is a leading Plasmodium falciparum blood-stage vaccine candidate. Another possible candidate, apical membrane antigen 1 (AMA1), was not efficacious in malaria-endemic populations, likely due to pre-existing antimalarial antibodies that interfered with the activity of vaccine-induced AMA1 antibodies, as judged by in vitro growth inhibition assay (GIA). To determine how pre-existing antibodies interact with vaccine-induced RH5 antibodies, we purify total and RH5-specific immunoglobulin Gs (IgGs) from malaria-exposed Malians and malaria-naive RH5 vaccinees. Infection-induced RH5 antibody titers are much lower than those induced by vaccination, and RH5-specific IgGs show differences in the binding site between the two populations. In GIA, Malian polyclonal IgGs show additive or synergistic interactions with RH5 human monoclonal antibodies and overall additive interactions with vaccine-induced polyclonal RH5 IgGs. These results suggest that pre-existing antibodies will interact favorably with vaccine-induced RH5 antibodies, in contrast to AMA1 antibodies. This study supports RH5 vaccine trials in malaria-endemic regions. RH5 IgG titers induced by infection are lower than those induced by RH5 vaccination Infection- and vaccine-induced RH5 IgGs have different specificity and avidity Infection- and vaccine-induced RH5 IgGs interact differently with RH5 mAbs Infection-induced IgGs generally do not reduce the activity of vaccine-induced IgGs
Collapse
Affiliation(s)
- Alexandra C Willcox
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Alex S Huber
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Jordan R Barrett
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Sarah E Silk
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - David Pulido
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Lloyd D W King
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Daniel G W Alanine
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Angela M Minassian
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Mahamadou Diakite
- Malaria Research and Training Center, Faculty of Medicine, Pharmacy, and Odontostomatology, University of Sciences, Techniques, and Technologies of Bamako, Bamako 1805, Mali
| | - Simon J Draper
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, United Kingdom
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| |
Collapse
|
16
|
Alonso S, Vidal M, Ruiz-Olalla G, González R, Jairoce C, Manaca MN, Vázquez-Santiago M, Balcells R, Vala A, Rupérez M, Cisteró P, Fuente-Soro L, Angov E, Coppel RL, Gamain B, Cavanagh D, Beeson JG, Nhacolo A, Sevene E, Aponte JJ, Macete E, Aguilar R, Mayor A, Menéndez C, Dobaño C, Moncunill G. HIV infection and placental malaria reduce maternal transfer of multiple antimalarial antibodies in Mozambican women. J Infect 2021; 82:45-57. [PMID: 33636218 DOI: 10.1016/j.jinf.2021.02.024] [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] [Received: 09/25/2020] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 10/22/2022]
Abstract
OBJECTIVES Maternal Plasmodium falciparum-specific antibodies may contribute to protect infants against severe malaria. Our main objective was to evaluate the impact of maternal HIV infection and placental malaria on the cord blood levels and efficiency of placental transfer of IgG and IgG subclasses. METHODS In a cohort of 341 delivering HIV-negative and HIV-positive mothers from southern Mozambique, we measured total IgG and IgG subclasses in maternal and cord blood pairs by quantitative suspension array technology against eight P. falciparum antigens: Duffy-binding like domains 3-4 of VAR2CSA from the erythrocyte membrane protein 1, erythrocyte-binding antigen 140, exported protein 1 (EXP1), merozoite surface proteins 1, 2 and 5, and reticulocyte-binding-homologue-4.2 (Rh4.2). We performed univariable and multivariable regression models to assess the association of maternal HIV infection, placental malaria, maternal variables and pregnancy outcomes on cord antibody levels and antibody transplacental transfer. RESULTS Maternal antibody levels were the main determinants of cord antibody levels. HIV infection and placental malaria reduced the transfer and cord levels of IgG and IgG1, and this was antigen-dependent. Low birth weight was associated with an increase of IgG2 in cord against EXP1 and Rh4.2. CONCLUSIONS We found lower maternally transferred antibodies in HIV-exposed infants and those born from mothers with placental malaria, which may underlie increased susceptibility to malaria in these children.
Collapse
Affiliation(s)
- Selena Alonso
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain
| | - Marta Vidal
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain
| | - Gemma Ruiz-Olalla
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain
| | - Raquel González
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - Chenjerai Jairoce
- Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - M Nelia Manaca
- Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - Miquel Vázquez-Santiago
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain
| | - Reyes Balcells
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - Anifa Vala
- Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - María Rupérez
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique; Present address: London School of Hygiene and Tropical Medicine (LSHTM). Keppel Street, WC1E 7HT, London, UK
| | - Pau Cisteró
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain
| | - Laura Fuente-Soro
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - Evelina Angov
- U.S. Military Malaria Vaccine Program, Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland, USA
| | - Ross L Coppel
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, VIC, Australia
| | - Benoit Gamain
- Université Sorbonne Paris Cité, Université Paris Diderot, Inserm, INTS, Unité Biologie Intégrée du Globule Rouge UMR_S1134, Laboratoire d'Excellence GR-Ex, Paris, France
| | - David Cavanagh
- Institute of Immunology & Infection Research and Centre for Immunity, Infection & Evolution, Ashworth Laboratories, School of Biological Sciences, University of Edinburgh, King's Buildings, Charlotte Auerbach Rd, Edinburgh, EH9 3FL, UK
| | | | - Arsenio Nhacolo
- Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - Esperança Sevene
- Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique; Department of Physiologic Science, Clinical Pharmacology, Faculty of Medicine, Eduardo Mondlane University, Maputo, Mozambique
| | - John J Aponte
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - Eusébio Macete
- Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - Ruth Aguilar
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain
| | - Alfredo Mayor
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - Clara Menéndez
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - Carlota Dobaño
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique.
| | - Gemma Moncunill
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique.
| |
Collapse
|
17
|
Ssewanyana I, Rek J, Rodriguez I, Wu L, Arinaitwe E, Nankabirwa JI, Beeson JG, Mayanja-Kizza H, Rosenthal PJ, Dorsey G, Kamya MR, Drakeley C, Greenhouse B, Tetteh KKA. Impact of a Rapid Decline in Malaria Transmission on Antimalarial IgG Subclasses and Avidity. Front Immunol 2021; 11:576663. [PMID: 33584643 PMCID: PMC7873448 DOI: 10.3389/fimmu.2020.576663] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 11/17/2020] [Indexed: 11/19/2022] Open
Abstract
Understanding how immunity to malaria is affected by declining transmission is important to aid vaccine design and understand disease resurgence. Both IgG subclasses and avidity of antigen-specific responses are important components of an effective immune response. Using a multiplex bead array assay, we measured the total IgG, IgG subclasses, and avidity profiles of responses to 18 P. falciparum blood stage antigens in samples from 160 Ugandans collected at two time points during high malaria transmission and two time points following a dramatic reduction in transmission. Results demonstrated that, for the antigens tested, (i) the rate of decay of total IgG following infection declined with age and was driven consistently by the decrease in IgG3 and occasionally the decrease in IgG1; (ii) the proportion of IgG3 relative to IgG1 in the absence of infection increased with age; (iii) the increase in avidity index (the strength of association between the antibody and antigen) following infection was largely due to a rapid loss of non-avid compared to avid total IgG; and (iv) both avid and non-avid total IgG in the absence of infection increased with age. Further studies are required to understand the functional differences between IgG1 and IgG3 in order to determine their contribution to the longevity of protective immunity to malaria. Measuring changes in antibody avidity may be a better approach of detecting affinity maturation compared to avidity index due to the differential expansion and contraction of high and low avidity total IgG.
Collapse
Affiliation(s)
- Isaac Ssewanyana
- Infectious Diseases Research Collaboration, Kampala, Uganda.,Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - John Rek
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Isabel Rodriguez
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Lindsey Wu
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Emmanuel Arinaitwe
- Infectious Diseases Research Collaboration, Kampala, Uganda.,Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Joaniter I Nankabirwa
- Infectious Diseases Research Collaboration, Kampala, Uganda.,School of Medicine, Makerere University, Kampala, Uganda
| | - James G Beeson
- Burnet Institute, Melbourne, VIC, Australia.,Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | | | - Philip J Rosenthal
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Grant Dorsey
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Moses R Kamya
- Infectious Diseases Research Collaboration, Kampala, Uganda.,School of Medicine, Makerere University, Kampala, Uganda
| | - Chris Drakeley
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Bryan Greenhouse
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States.,Chan Zuckerberg Biohub, San Francisco, CA, United States
| | - Kevin K A Tetteh
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
18
|
Acquisition and decay of IgM and IgG responses to merozoite antigens after Plasmodium falciparum malaria in Ghanaian children. PLoS One 2020; 15:e0243943. [PMID: 33332459 PMCID: PMC7746192 DOI: 10.1371/journal.pone.0243943] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/30/2020] [Indexed: 12/18/2022] Open
Abstract
Developing a vaccine against Plasmodium falciparum malaria has been challenging, primarily due to high levels of antigen polymorphism and a complex parasite lifecycle. Immunization with the P. falciparum merozoite antigens PfMSRP5, PfSERA9, PfRAMA, PfCyRPA and PfRH5 has been shown to give rise to growth inhibitory and synergistic antisera. Therefore, these five merozoite proteins are considered to be promising candidates for a second-generation multivalent malaria vaccine. Nevertheless, little is known about IgG and IgM responses to these antigens in populations that are naturally exposed to P. falciparum. In this study, serum samples from clinically immune adults and malaria exposed children from Ghana were studied to compare levels of IgG and IgM specific for PfMSRP5, PfSERA9, PfRAMA, PfCyRPA and PfRH5. All five antigens were found to be specifically recognized by both IgM and IgG in serum from clinically immune adults and from children with malaria. Longitudinal analysis of the latter group showed an early, transient IgM response that was followed by IgG, which peaked 14 days after the initial diagnosis. IgG levels and parasitemia did not correlate, whereas parasitemia was weakly positively correlated with IgM levels. These findings show that IgG and IgM specific for merozoite antigens PfMSRP5, PfSERA9, PfRAMA, PfCyRPA and PfRH5 are high in children during P. falciparum malaria, but that the IgM induction and decline occurs earlier in infection than that of IgG.
Collapse
|
19
|
Ezoe S, Palacpac NMQ, Tetsutani K, Yamamoto K, Okada K, Taira M, Nishida S, Hirata H, Ogata A, Yamada T, Yagi M, Edula JR, Oishi Y, Tougan T, Ishii KJ, Myoui A, Horii T. First-in-human randomised trial and follow-up study of Plasmodium falciparum blood-stage malaria vaccine BK-SE36 with CpG-ODN(K3). Vaccine 2020; 38:7246-7257. [PMID: 33012605 DOI: 10.1016/j.vaccine.2020.09.056] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND BK-SE36 is blood-stage malaria vaccine candidate that is undergoing clinical trials. Here, the safety and immunogenicity of BK-SE36 with a novel adjuvant, CpG-ODN(K3) (thus, BK-SE36/CpG) was assessed in a phase 1a trial in Japan. METHODS An investigator-initiated, randomised, single-blind, placebo-controlled, dose-escalation study was conducted at Osaka University Hospital with 26 healthy malaria naïve Japanese male adults. The trial was conducted in two stages: Stage/Group 1, half-dose (n = 7 for BK-SE36/CpG and n = 3 for control) and Stage/Group 2, full-dose (n = 11 for BK-SE36/CpG and n = 5 for control). There were two intramuscular vaccinations 21 days apart for both half-dose (0.5 ml: 50 µg SE36 + 500 µg aluminum + 500 µg K3) and full-dose (1.0 ml: 100 µg SE36 + 1000 µg aluminum + 1000 µg K3). A one-year follow-up was done to monitor changes in autoimmune markers and vaccine-induced antibody response. RESULTS BK-SE36/CpG was well tolerated. Vaccination site reactions were similar to those observed with BK-SE36. During the trial and follow-up period, no subject had clinical evidence of autoimmune disease. The full-dose group had significantly higher titres than the half-dose group (Student's t-test, p = 0.002) at 21 days post-second vaccination. Antibody titres remained above baseline values during 12 months of follow-up. The vaccine induced antibody was mostly composed of IgG1 and IgM, and recognised epitopes close to the polyserine region located in the middle of SE36. CONCLUSIONS BK-SE36/CpG has an acceptable safety profile. Use of CpG-ODN(K3) greatly enhanced immunogenicity in malaria naïve Japanese adults when compared to BK-SE36 alone. The utility of BK-SE36/CpG is currently under evaluation in a malaria endemic setting in West Africa. TRIAL REGISTRATION JMACCT Clinical Trial Registry JMA-IIA00109.
Collapse
Affiliation(s)
- Sachiko Ezoe
- Medical Center for Translational Research, Department of Medical Innovation, Osaka University Hospital, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Environmental Space Infection Control, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Nirianne Marie Q Palacpac
- Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kohhei Tetsutani
- Laboratory of Adjuvant Innovation/Mockup Vaccine, Center for Vaccine Adjuvant Research, National Institute of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki City, Osaka 567-0085, Japan; Laboratory of Vaccine Science, Immunology Frontier Research Center (IFReC), Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kouji Yamamoto
- Data Coordinating Center, Department of Medical Innovation, Osaka University Hospital, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kiyoshi Okada
- Department of Orthopaedics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masaki Taira
- Medical Center for Translational Research, Department of Medical Innovation, Osaka University Hospital, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Sumiyuki Nishida
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Haruhiko Hirata
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Atsushi Ogata
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tomomi Yamada
- Data Coordinating Center, Department of Medical Innovation, Osaka University Hospital, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masanori Yagi
- Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Jyotheeswara R Edula
- Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yuko Oishi
- Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takahiro Tougan
- Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ken J Ishii
- Laboratory of Adjuvant Innovation/Mockup Vaccine, Center for Vaccine Adjuvant Research, National Institute of Biomedical Innovation, Health and Nutrition, 7-6-8, Saito-Asagi, Ibaraki City, Osaka 567-0085, Japan; Laboratory of Vaccine Science, Immunology Frontier Research Center (IFReC), Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Akira Myoui
- Medical Center for Translational Research, Department of Medical Innovation, Osaka University Hospital, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Toshihiro Horii
- Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
20
|
Bemani P, Amirghofran Z, Mohammadi M. Designing a multi-epitope vaccine against blood-stage of Plasmodium falciparum by in silico approaches. J Mol Graph Model 2020; 99:107645. [PMID: 32454399 DOI: 10.1016/j.jmgm.2020.107645] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/11/2022]
Abstract
Plasmodium falciparum causes the most severe form of malaria disease and is the major cause of infection-related mortalities in the world. Due to increasing in P. falciparum resistance to the first-line antimalarial drugs, an effective vaccine for the control and elimination of malaria infection is urgent. Because the pathogenesis of malaria disease results from blood-stage infection, and all of the symptoms and clinical illness of malaria occur during this stage, there is a strong rationale to develop vaccine against this stage. In the present study, different structural-vaccinology and immuno informatics tools were applied to design an effective antibody-inducing multi-epitope vaccine against the blood-stage of P. falciparum. The designed multi-epitope vaccine was composed of three main parts including B cell epitopes, T helper (Th) cell epitopes, and two adjuvant motives (HP91 and RS09), which were linked to each other via proper linkers. B cell and T cell epitopes were derived from four protective antigens expressed on the surface of merozoites, which are critical to invade the erythrocytes. HP91 and RS09 adjuvants and Th cell epitopes were used to induce, enhance and direct the best form of humoral immune-response against P. falciparum surface merozoite antigens. The vaccine construct was modeled, and after model quality evaluation and refinement by different software, the high-quality 3D-structure model of the vaccine was achieved. Analysis of immunological and physicochemical features of the vaccine showed acceptable results. We believe that this multi-epitope vaccine can be effective for preventing malaria disease caused by P. falciparum.
Collapse
Affiliation(s)
- Peyman Bemani
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Zahra Amirghofran
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mozafar Mohammadi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Acquah FK, Lo AC, Akyea-Mensah K, Abagna HB, Faye B, Theisen M, Gyan BA, Amoah LE. Stage-specific Plasmodium falciparum immune responses in afebrile adults and children living in the Greater Accra Region of Ghana. Malar J 2020; 19:64. [PMID: 32041620 PMCID: PMC7011432 DOI: 10.1186/s12936-020-3146-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/30/2020] [Indexed: 12/02/2022] Open
Abstract
Background Asymptomatic carriage of Plasmodium falciparum is widespread in adults and children living in malaria-endemic countries. This study identified the prevalence of malaria parasites and the corresponding levels of naturally acquired anti-parasite antibody levels in afebrile adults living in two communities in the Greater Accra Region of Ghana. Methods Two cross-sectional studies conducted in January and February 2016 and repeated in July and August 2016 recruited subjects aged between 6 and 75 years from high parasite prevalence (Obom) and low parasite prevalence (Asutsuare) communities. Whole blood (5 ml) was collected from each volunteer, plasma was aliquoted and frozen until needed. An aliquot (10 µl) of the blood was used to prepare thick and thin blood smears, 100 µl was preserved in Trizol and the rest was separated into plasma and blood cells and each stored at − 20 °C until needed. Anti-MSP3 and Pfs230 antibody levels were measured using ELISA. Results Asexual parasite and gametocyte prevalence were higher in Obom than Asutsuare. Antibody (IgG, IgG1, IgG3, IgM) responses against the asexual parasite antigen MSP3 and gametocyte antigen Pfs230 were higher in Obom during the course of the study except for IgM responses against Pfs230, which was higher in Asutsuare than in Obom during the rainy season. Antibody responses in Asutsuare were more significantly associated with age than the responses measured in Obom. Conclusion The pattern of antibody responses measured in people living in the high and low malaria transmission setting was similar. All antibody responses measured against the asexual antigen MSP3 increased, however, IgG and IgG1 responses against gametocyte antigen Pfs230 decreased in moving from the dry to the peak season in both sites. Whilst asexual and gametocyte prevalence was similar between the seasons in the low transmission setting, in the high transmission setting asexual parasite prevalence increased but gametocyte prevalence decreased in the rainy season relative to the dry season.
Collapse
Affiliation(s)
- Festus K Acquah
- Immunology Department, Noguchi Memorial Institute for Medical Research (NMIMR), University of Ghana, Accra, Ghana.,West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
| | - Aminata C Lo
- Immunology Department, Noguchi Memorial Institute for Medical Research (NMIMR), University of Ghana, Accra, Ghana.,Parasitology Department, University Cheikh Anta Diop, Dakar, Senegal
| | - Kwadwo Akyea-Mensah
- Immunology Department, Noguchi Memorial Institute for Medical Research (NMIMR), University of Ghana, Accra, Ghana
| | - Hamza B Abagna
- Immunology Department, Noguchi Memorial Institute for Medical Research (NMIMR), University of Ghana, Accra, Ghana
| | - Babacar Faye
- Parasitology Department, University Cheikh Anta Diop, Dakar, Senegal
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Ben A Gyan
- Immunology Department, Noguchi Memorial Institute for Medical Research (NMIMR), University of Ghana, Accra, Ghana
| | - Linda E Amoah
- Immunology Department, Noguchi Memorial Institute for Medical Research (NMIMR), University of Ghana, Accra, Ghana. .,West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana.
| |
Collapse
|
22
|
Garzón-Ospina D, Buitrago SP. Igh locus structure and evolution in Platyrrhines: new insights from a genomic perspective. Immunogenetics 2019; 72:165-179. [PMID: 31838542 DOI: 10.1007/s00251-019-01151-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/20/2019] [Indexed: 12/17/2022]
Abstract
Non-human primates have been used as animal models because of their phylogenetic closeness to humans. However, the genetic differences between humans and non-human primates must be considered to select the appropriate animal models. Recently, New World monkeys (Platyrrhines) have generated a higher interest in biomedical research, especially in assessing vaccine safety and immunogenicity. Given the continued and renewed interest in Platyrrhines as biomedical models, it is a necessary to have a better and more complete understanding of their immune system and its implications for research. Immunoglobulins (Ig) are the main proteins that mediate humoral immunity. These proteins have evolved as part of an adaptive immune response system derived from ancient vertebrates. There are at least four Ig classes in Prosimians, whereas five have been reported in Catarrhines. Information on the structure and evolution of the loci containing immunoglobulin heavy chain constant genes (Igh) in Platyrrhines, however, is limited. Here, Igh loci were characterized in 10 Platyrrhines using the available whole genome sequences. Human and Macaca Igh loci were also assessed to compare them with their Platyrrhines counterparts. Differences in Igh locus structure were observed between Platyrrhines and Catarrhines. Noteworthy changes occur in the γ gene, which encodes a key Ig involved in organism defense that would favor protection after vaccination. The remarkable differences between the immunoglobulin proteins of Platyrrhines and Catarrhines warrant a cautionary message to biomedical researchers.
Collapse
Affiliation(s)
- Diego Garzón-Ospina
- Pgame - Population Genetics And Molecular Evolution, Fundación Scient, Carrera 16-3 # 35-41, Tunja, Boyacá, Colombia.
| | - Sindy P Buitrago
- Pgame - Population Genetics And Molecular Evolution, Fundación Scient, Carrera 16-3 # 35-41, Tunja, Boyacá, Colombia.
| |
Collapse
|
23
|
Tentokam BCN, Amaratunga C, Alani NAH, MacDonald NJ, Narum DL, Salinas ND, Kwan JL, Suon S, Sreng S, Pereira DB, Tolia NH, Fujiwara RT, Bueno LL, Duffy PE, Coelho CH. Naturally Acquired Antibody Response to Malaria Transmission Blocking Vaccine Candidate Pvs230 Domain 1. Front Immunol 2019; 10:2295. [PMID: 31636633 PMCID: PMC6788386 DOI: 10.3389/fimmu.2019.02295] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/11/2019] [Indexed: 01/27/2023] Open
Abstract
Plasmodium vivax malaria incidence has increased in Latin America and Asia and is responsible for nearly 74.1% of malaria cases in Latin America. Immune responses to P. vivax are less well characterized than those to P. falciparum, partly because P. vivax is more difficult to cultivate in the laboratory. While antibodies are known to play an important role in P. vivax disease control, few studies have evaluated responses to P. vivax sexual stage antigens. We collected sera or plasma samples from P. vivax-infected subjects from Brazil (n = 70) and Cambodia (n = 79) to assess antibody responses to domain 1 of the gametocyte/gamete stage protein Pvs230 (Pvs230D1M). We found that 27.1% (19/70) and 26.6% (21/79) of subjects from Brazil and Cambodia, respectively, presented with detectable antibody responses to Pvs230D1M antigen. The most frequent subclasses elicited in response to Pvs230D1M were IgG1 and IgG3. Although age did not correlate significantly with Pvs230D1M antibody levels overall, we observed significant differences between age strata. Hemoglobin concentration inversely correlated with Pvs230D1M antibody levels in Brazil, but not in Cambodia. Additionally, we analyzed the antibody response against Pfs230D1M, the P. falciparum ortholog of Pvs230D1M. We detected antibodies to Pfs230D1M in 7.2 and 16.5% of Brazilian and Cambodian P. vivax-infected subjects. Depletion of Pvs230D1M IgG did not impair the response to Pfs230D1M, suggesting pre-exposure to P. falciparum, or co-infection. We also analyzed IgG responses to sporozoite protein PvCSP (11.4 and 41.8% in Brazil and Cambodia, respectively) and to merozoite protein PvDBP-RII (67.1 and 48.1% in Brazil and Cambodia, respectively), whose titers also inversely correlated with hemoglobin concentration only in Brazil. These data establish patterns of seroreactivity to sexual stage Pvs230D1M and show similar antibody responses among P. vivax-infected subjects from regions of differing transmission intensity in Brazil and Cambodia.
Collapse
Affiliation(s)
- Bergeline C Nguemwo Tentokam
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Chanaki Amaratunga
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Rockville, MD, United States
| | - Nada A H Alani
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Nicholas J MacDonald
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - David L Narum
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Nichole D Salinas
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Jennifer L Kwan
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Seila Suon
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Sokunthea Sreng
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | | | - Niraj H Tolia
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Ricardo T Fujiwara
- Department of Parasitology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Lilian L Bueno
- Department of Parasitology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Camila H Coelho
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
24
|
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: 33] [Impact Index Per Article: 5.5] [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
|
25
|
Humoral immunity prevents clinical malaria during Plasmodium relapses without eliminating gametocytes. PLoS Pathog 2019; 15:e1007974. [PMID: 31536608 PMCID: PMC6752766 DOI: 10.1371/journal.ppat.1007974] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 07/10/2019] [Indexed: 12/19/2022] Open
Abstract
Plasmodium relapses are attributed to the activation of dormant liver-stage parasites and are responsible for a significant number of recurring malaria blood-stage infections. While characteristic of human infections caused by P. vivax and P. ovale, their relative contribution to malaria disease burden and transmission remains poorly understood. This is largely because it is difficult to identify ‘bona fide’ relapse infections due to ongoing transmission in most endemic areas. Here, we use the P. cynomolgi–rhesus macaque model of relapsing malaria to demonstrate that clinical immunity can form after a single sporozoite-initiated blood-stage infection and prevent illness during relapses and homologous reinfections. By integrating data from whole blood RNA-sequencing, flow cytometry, P. cynomolgi-specific ELISAs, and opsonic phagocytosis assays, we demonstrate that this immunity is associated with a rapid recall response by memory B cells that expand and produce anti-parasite IgG1 that can mediate parasite clearance of relapsing parasites. The reduction in parasitemia during relapses was mirrored by a reduction in the total number of circulating gametocytes, but importantly, the cumulative proportion of gametocytes increased during relapses. Overall, this study reveals that P. cynomolgi relapse infections can be clinically silent in macaques due to rapid memory B cell responses that help to clear asexual-stage parasites but still carry gametocytes. Plasmodium vivax contributes significantly to global malaria morbidity and remains a major obstacle for malaria elimination due to its ability to form dormant stages in the liver. These forms can become activated to cause relapsing blood-stage infections. Relapses remain poorly understood because it is difficult to verify whether P. vivax blood-stage infections in patients are due to new infections or relapses in most cases. Here, we use a nonhuman primate model of Plasmodium vivax malaria in concert with state-of-the-art immunological and molecular techniques to assess pathogenesis, host responses, and circulating gametocyte levels during relapses. We found that relapses were clinically silent compared to initial infections, and they were associated with a robust memory B cell response. This response resulted in the production of antibodies that were able to mediate clearance of asexual parasites. Despite this rapid immune protection, the sexual-stage gametocytes continued to circulate. Our study provides mechanistic insights into the host-parasite interface during Plasmodium relapse infections and demonstrates that clinically silent relapses can harbor gametocytes that may be infectious to mosquitoes.
Collapse
|
26
|
Healer J, Wong W, Thompson JK, He W, Birkinshaw RW, Miura K, Long CA, Soroka V, Søgaard TMM, Jørgensen T, de Jongh WA, Weir C, Svahn E, Czabotar PE, Tham W, Mueller I, Barlow PN, Cowman AF. Neutralising antibodies block the function of Rh5/Ripr/CyRPA complex during invasion of Plasmodium falciparum into human erythrocytes. Cell Microbiol 2019; 21:e13030. [PMID: 30965383 PMCID: PMC6594224 DOI: 10.1111/cmi.13030] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/04/2019] [Accepted: 03/11/2019] [Indexed: 01/19/2023]
Abstract
An effective vaccine is a priority for malaria control and elimination. The leading candidate in the Plasmodium falciparum blood stage is PfRh5. PfRh5 assembles into trimeric complex with PfRipr and PfCyRPA in the parasite, and this complex is essential for erythrocyte invasion. In this study, we show that antibodies specific for PfRh5 and PfCyRPA prevent trimeric complex formation. We identify the EGF-7 domain on PfRipr as a neutralising epitope and demonstrate that antibodies against this region act downstream of complex formation to prevent merozoite invasion. Antibodies against the C-terminal region of PfRipr were more inhibitory than those against either PfRh5 or PfCyRPA alone, and a combination of antibodies against PfCyRPA and PfRipr acted synergistically to reduce invasion. This study supports prioritisation of PfRipr for development as part of a next-generation antimalarial vaccine.
Collapse
Affiliation(s)
- Julie Healer
- Infection and ImmunityWalter and Eliza Hall Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneMelbourneVictoriaAustralia
| | - Wilson Wong
- Infection and ImmunityWalter and Eliza Hall Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneMelbourneVictoriaAustralia
| | - Jennifer K. Thompson
- Infection and ImmunityWalter and Eliza Hall Institute of Medical ResearchMelbourneVictoriaAustralia
| | - Wengqiang He
- Infection and ImmunityWalter and Eliza Hall Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneMelbourneVictoriaAustralia
| | - Richard W. Birkinshaw
- Infection and ImmunityWalter and Eliza Hall Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneMelbourneVictoriaAustralia
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector ResearchNational Institute of Allergy and Infectious Diseases, National Institutes of HealthBethesdaMarylandUSA
| | - Carol A. Long
- Laboratory of Malaria and Vector ResearchNational Institute of Allergy and Infectious Diseases, National Institutes of HealthBethesdaMarylandUSA
| | | | | | | | | | - Christopher Weir
- Infection and ImmunityWalter and Eliza Hall Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneMelbourneVictoriaAustralia
- Schools of Chemistry and Biological SciencesUniversity of EdinburghEdinburghScotland, UK
| | - Ella Svahn
- Schools of Chemistry and Biological SciencesUniversity of EdinburghEdinburghScotland, UK
| | - Peter E. Czabotar
- Infection and ImmunityWalter and Eliza Hall Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneMelbourneVictoriaAustralia
| | - Wai‐Hong Tham
- Infection and ImmunityWalter and Eliza Hall Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneMelbourneVictoriaAustralia
| | - Ivo Mueller
- Infection and ImmunityWalter and Eliza Hall Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneMelbourneVictoriaAustralia
| | - Paul N. Barlow
- Schools of Chemistry and Biological SciencesUniversity of EdinburghEdinburghScotland, UK
| | - Alan F. Cowman
- Infection and ImmunityWalter and Eliza Hall Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
27
|
Fan Y, Mu Y, Lu L, Tian Y, Yuan F, Zhou B, Yu C, Wang Z, Li X, Lei S, Xu Y, Wu D, Yang L. Hydrogen peroxide-inactivated bacteria induces potent humoral and cellular immune responses and releases nucleic acids. Int Immunopharmacol 2019; 69:389-397. [DOI: 10.1016/j.intimp.2019.01.055] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/24/2019] [Accepted: 01/31/2019] [Indexed: 01/07/2023]
|
28
|
Dobaño C, Santano R, Vidal M, Jiménez A, Jairoce C, Ubillos I, Dosoo D, Aguilar R, Williams NA, Díez-Padrisa N, Ayestaran A, Valim C, Asante KP, Owusu-Agyei S, Lanar D, Chauhan V, Chitnis C, Dutta S, Angov E, Gamain B, Coppel RL, Beeson JG, Reiling L, Gaur D, Cavanagh D, Gyan B, Nhabomba AJ, Campo JJ, Moncunill G. Differential Patterns of IgG Subclass Responses to Plasmodium falciparum Antigens in Relation to Malaria Protection and RTS,S Vaccination. Front Immunol 2019; 10:439. [PMID: 30930896 PMCID: PMC6428712 DOI: 10.3389/fimmu.2019.00439] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 02/19/2019] [Indexed: 12/24/2022] Open
Abstract
Naturally acquired immunity (NAI) to Plasmodium falciparum malaria is mainly mediated by IgG antibodies but the subclasses, epitope targets and effector functions have not been unequivocally defined. Dissecting the type and specificity of antibody responses mediating NAI is a key step toward developing more effective vaccines to control the disease. We investigated the role of IgG subclasses to malaria antigens in protection against disease and the factors that affect their levels, including vaccination with RTS,S/AS01E. We analyzed plasma and serum samples at baseline and 1 month after primary vaccination with RTS,S or comparator in African children and infants participating in a phase 3 trial in two sites of different malaria transmission intensity: Kintampo in Ghana and Manhiça in Mozambique. We used quantitative suspension array technology (qSAT) to measure IgG1−4 responses to 35 P. falciparum pre-erythrocytic and blood stage antigens. Our results show that the pattern of IgG response is predominantly IgG1 or IgG3, with lower levels of IgG2 and IgG4. Age, site and RTS,S vaccination significantly affected antibody subclass levels to different antigens and susceptibility to clinical malaria. Univariable and multivariable analysis showed associations with protection mainly for cytophilic IgG3 levels to selected antigens, followed by IgG1 levels and, unexpectedly, also with IgG4 levels, mainly to antigens that increased upon RTS,S vaccination such as MSP5 and MSP1 block 2, among others. In contrast, IgG2 was associated with malaria risk. Stratified analysis in RTS,S vaccinees pointed to novel associations of IgG4 responses with immunity mainly involving pre-erythrocytic antigens upon RTS,S vaccination. Multi-marker analysis revealed a significant contribution of IgG3 responses to malaria protection and IgG2 responses to malaria risk. We propose that the pattern of cytophilic and non-cytophilic IgG antibodies is antigen-dependent and more complex than initially thought, and that mechanisms of both types of subclasses could be involved in protection. Our data also suggests that RTS,S efficacy is significantly affected by NAI, and indicates that RTS,S vaccination significantly alters NAI.
Collapse
Affiliation(s)
- Carlota Dobaño
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde de Manhiça (CISM), Manhiça, Mozambique
| | - Rebeca Santano
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Marta Vidal
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Alfons Jiménez
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Spanish Consortium for Research in Epidemiology and Public Health (CIBERESP), Barcelona, Spain
| | - Chenjerai Jairoce
- Centro de Investigação em Saúde de Manhiça (CISM), Manhiça, Mozambique
| | - Itziar Ubillos
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - David Dosoo
- Kintampo Health Research Centre, Kintampo, Ghana
| | - Ruth Aguilar
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Nana Aba Williams
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | | | | | - Clarissa Valim
- Department of Osteopathic Medical Specialties, Michigan State University, East Lansing, MI, United States.,Department of Immunology and Infectious Diseases, Harvard T.H. Chen School of Public Health, Boston, MA, United States
| | | | - Seth Owusu-Agyei
- Kintampo Health Research Centre, Kintampo, Ghana.,Disease Control Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - David Lanar
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Virander Chauhan
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Chetan Chitnis
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Sheetij Dutta
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Evelina Angov
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Benoit Gamain
- Unité Biologie Intégrée du Globule Rouge, Laboratoire d'Excellence GR-Ex, UMR_S1134, Inserm, INTS, Université Sorbonne Paris Cité, Université Paris Diderot, Paris, France
| | - Ross L Coppel
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne, VIC, Australia
| | | | | | - Deepak Gaur
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India.,Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - David Cavanagh
- Ashworth Laboratories, Centre for Immunity, Infection and Evolution, School of Biological Sciences, Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Ben Gyan
- Kintampo Health Research Centre, Kintampo, Ghana.,Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | | | - Joseph J Campo
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde de Manhiça (CISM), Manhiça, Mozambique
| | - Gemma Moncunill
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde de Manhiça (CISM), Manhiça, Mozambique
| |
Collapse
|
29
|
Kurtovic L, Agius PA, Feng G, Drew DR, Ubillos I, Sacarlal J, Aponte JJ, Fowkes FJI, Dobaño C, Beeson JG. Induction and decay of functional complement-fixing antibodies by the RTS,S malaria vaccine in children, and a negative impact of malaria exposure. BMC Med 2019; 17:45. [PMID: 30798787 PMCID: PMC6388494 DOI: 10.1186/s12916-019-1277-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/31/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Leading malaria vaccine, RTS,S, is based on the circumsporozoite protein (CSP) of sporozoites. RTS,S confers partial protection against malaria in children, but efficacy wanes relatively quickly after primary immunization. Vaccine efficacy has some association with anti-CSP IgG; however, it is unclear how these antibodies function, and how functional antibodies are induced and maintained over time. Recent studies identified antibody-complement interactions as a potentially important immune mechanism against sporozoites. Here, we investigated whether RTS,S vaccine-induced antibodies could function by interacting with complement. METHODS Serum samples were selected from children in a phase IIb trial of RTS,S/AS02A conducted at two study sites of high and low malaria transmission intensity in Manhiça, Mozambique. Samples following primary immunization and 5-year post-immunization follow-up time points were included. Vaccine-induced antibodies were characterized by isotype, subclass, and epitope specificity, and tested for the ability to fix and activate complement. We additionally developed statistical methods to model the decay and determinants of functional antibodies after vaccination. RESULTS RTS,S vaccination induced anti-CSP antibodies that were mostly IgG1, with some IgG3, IgG2, and IgM. Complement-fixing antibodies were effectively induced by vaccination, and targeted the central repeat and C-terminal regions of CSP. Higher levels of complement-fixing antibodies were associated with IgG that equally recognized both the central repeat and C-terminal regions of CSP. Older age and higher malaria exposure were significantly associated with a poorer induction of functional antibodies. There was a marked decay in functional complement-fixing antibodies within months after vaccination, as well as decays in IgG subclasses and IgM. Statistical modeling suggested the decay in complement-fixing antibodies was mostly attributed to the waning of anti-CSP IgG1, and to a lesser extent IgG3. CONCLUSIONS We demonstrate for the first time that RTS,S can induce complement-fixing antibodies in young malaria-exposed children. The short-lived nature of functional responses mirrors the declining vaccine efficacy of RTS,S over time. The negative influence of age and malaria exposure on functional antibodies has implications for understanding vaccine efficacy in different settings. These findings provide insights into the mechanisms and longevity of vaccine-induced immunity that will help inform the future development of highly efficacious and long-lasting malaria vaccines.
Collapse
Affiliation(s)
- Liriye Kurtovic
- Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia
| | - Paul A Agius
- Burnet Institute, Melbourne, Australia.,Department of Epidemiology and Preventative Medicine, Monash University, Melbourne, Australia
| | | | | | - Itziar Ubillos
- ISGlobal, Hospital Clínic Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Jahit Sacarlal
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique.,Faculdade de Medicina, Universidade Eduardo Mondlane (UEM), Maputo, Mozambique
| | - John J Aponte
- ISGlobal, Hospital Clínic Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
| | - Freya J I Fowkes
- Burnet Institute, Melbourne, Australia.,Department of Epidemiology and Preventative Medicine, Monash University, Melbourne, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - Carlota Dobaño
- ISGlobal, Hospital Clínic Universitat de Barcelona, Barcelona, Catalonia, Spain.,Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
| | - James G Beeson
- Burnet Institute, Melbourne, Australia. .,Department of Immunology and Pathology, Monash University, Melbourne, Australia. .,Department of Microbiology, Monash University, Clayton, Australia. .,Department of Medicine, The University of Melbourne, Parkville, Australia.
| |
Collapse
|
30
|
Damelang T, Rogerson SJ, Kent SJ, Chung AW. Role of IgG3 in Infectious Diseases. Trends Immunol 2019; 40:197-211. [PMID: 30745265 DOI: 10.1016/j.it.2019.01.005] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 01/14/2019] [Accepted: 01/14/2019] [Indexed: 12/20/2022]
Abstract
IgG3 comprises only a minor fraction of IgG and has remained relatively understudied until recent years. Key physiochemical characteristics of IgG3 include an elongated hinge region, greater molecular flexibility, extensive polymorphisms, and additional glycosylation sites not present on other IgG subclasses. These characteristics make IgG3 a uniquely potent immunoglobulin, with the potential for triggering effector functions including complement activation, antibody (Ab)-mediated phagocytosis, or Ab-mediated cellular cytotoxicity (ADCC). Recent studies underscore the importance of IgG3 effector functions against a range of pathogens and have provided approaches to overcome IgG3-associated limitations, such as allotype-dependent short Ab half-life, and excessive proinflammatory activation. Understanding the molecular and functional properties of IgG3 may facilitate the development of improved Ab-based immunotherapies and vaccines against infectious diseases.
Collapse
Affiliation(s)
- Timon Damelang
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Stephen J Rogerson
- Department of Medicine, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Amy W Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
31
|
Targets of complement-fixing antibodies in protective immunity against malaria in children. Nat Commun 2019; 10:610. [PMID: 30723225 PMCID: PMC6363798 DOI: 10.1038/s41467-019-08528-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/07/2019] [Indexed: 12/31/2022] Open
Abstract
Antibodies against P. falciparum merozoites fix complement to inhibit blood-stage replication in naturally-acquired and vaccine-induced immunity; however, specific targets of these functional antibodies and their importance in protective immunity are unknown. Among malaria-exposed individuals, we show that complement-fixing antibodies to merozoites are more strongly correlated with protective immunity than antibodies that inhibit growth quantified using the current reference assay for merozoite vaccine evaluation. We identify merozoite targets of complement-fixing antibodies and identify antigen-specific complement-fixing antibodies that are strongly associated with protection from malaria in a longitudinal study of children. Using statistical modelling, combining three different antigens targeted by complement-fixing antibodies could increase the potential protective effect to over 95%, and we identify antigens that were common in the most protective combinations. Our findings support antibody-complement interactions against merozoite antigens as important anti-malaria immune mechanisms, and identify specific merozoite antigens for further evaluation as vaccine candidates. Antibodies against Plasmodium falciparum merozoites that fix complement can inhibit blood-stage replication. Here, Reiling et al. show that complement-fixing antibodies strongly correlate with protective immunity in children, identify the merozoite targets, and predict antigen combinations that should result in strong protection.
Collapse
|
32
|
Yman V, White MT, Asghar M, Sundling C, Sondén K, Draper SJ, Osier FHA, Färnert A. Antibody responses to merozoite antigens after natural Plasmodium falciparum infection: kinetics and longevity in absence of re-exposure. BMC Med 2019; 17:22. [PMID: 30696449 PMCID: PMC6352425 DOI: 10.1186/s12916-019-1255-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 01/08/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Antibodies against merozoite antigens are key components of malaria immunity. The naturally acquired antibody response to these antigens is generally considered short-lived; however, the underlying mechanisms remain unclear. Prospective studies of travellers with different levels of prior exposure, returning to malaria-free countries with Plasmodium infection, offer a unique opportunity to investigate the kinetics and composition of the antibody response after natural infection. METHODS Adults diagnosed with P. falciparum malaria in Stockholm, Sweden (20 likely malaria naïve and 41 with repeated previous exposure during residency in sub-Saharan Africa) were sampled at diagnosis and 10 days and 1, 3, 6, and 12 months after treatment. Total and subclass-specific IgG responses to P. falciparum merozoite antigens (AMA-1, MSP-119, MSP-2, MSP-3, and RH5) and tetanus toxoid were measured by multiplex bead-based immunoassays and ELISA. Mathematical modelling was used to estimate the exposure-dependent longevity of antibodies and antibody-secreting cells (ASCs). RESULTS A majority of individuals mounted detectable antibody responses towards P. falciparum merozoite antigens at diagnosis; however, the magnitude and breadth were greater in individuals with prior exposure. In both exposure groups, antibody levels increased rapidly for 2 weeks and decayed thereafter. Previously exposed individuals maintained two- to ninefold greater antibody levels throughout the 1-year follow-up. The half-lives of malaria-specific long-lived ASCs, responsible for maintaining circulating antibodies, ranged from 1.8 to 3.7 years for merozoite antigens and were considerably short compared to tetanus-specific ASCs. Primary infected individuals did acquire a long-lived component of the antibody response; however, the total proportion of long-lived ASCs generated in response to infection was estimated not to exceed 10%. In contrast, previously exposed individuals maintained substantially larger numbers of long-lived ASCs (10-56% of total ASCs). CONCLUSION The short-lived nature of the naturally acquired antibody response, to all tested merozoite antigens, following primary malaria infection can be attributed to a combination of a poor acquisition and short half-life of long-lived ASCs. Greater longevity is acquired with repeated infections and can be explained by the maintenance of larger numbers of long-lived ASCs. These insights advance our understanding of naturally acquired malaria immunity and will guide strategies for further development of both vaccines and serological tools to monitor exposure.
Collapse
Affiliation(s)
- Victor Yman
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 76, Stockholm, Sweden.
| | - Michael T White
- Department of Parasites and Insect Vectors, Institut Pasteur, 25-28 Rue du Dr Roux, 75015, Paris, France
| | - Muhammad Asghar
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Christopher Sundling
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Klara Sondén
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 76, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Simon J Draper
- Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Faith H A Osier
- Kenya Medical Research Institute - Wellcome Trust Research Program, Centre for Geographic Medicine Research-Coast, PO Box 230-80108, Kilifi, Kenya
- Centre for Infectious Diseases, Parasitology, Heidelberg University Hospital, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Anna Färnert
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 76, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, 171 76, Stockholm, Sweden
| |
Collapse
|
33
|
Wilson KL, Flanagan KL, Prakash MD, Plebanski M. Malaria vaccines in the eradication era: current status and future perspectives. Expert Rev Vaccines 2019; 18:133-151. [PMID: 30601095 DOI: 10.1080/14760584.2019.1561289] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The challenge to eradicate malaria is an enormous task that will not be achieved by current control measures, thus an efficacious and long-lasting malaria vaccine is required. The licensing of RTS, S/AS01 is a step forward in providing some protection, but a malaria vaccine that protects across multiple transmission seasons is still needed. To achieve this, inducing beneficial immune responses while minimising deleterious non-targeted effects will be essential. AREAS COVERED This article discusses the current challenges and advances in malaria vaccine development and reviews recent human clinical trials for each stage of infection. Pubmed and ScienceDirect were searched, focusing on cell mediated immunity and how T cell subsets might be targeted in future vaccines using novel adjuvants and emerging vaccine technologies. EXPERT COMMENTARY Despite decades of research there is no highly effective licensed malaria vaccine. However, there is cause for optimism as new adjuvants and vaccine systems emerge, and our understanding of correlates of protection increases, especially regarding cellular immunity. The new field of heterologous (non-specific) effects of vaccines also highlights the broader consequences of immunization. Importantly, the WHO led Malaria Vaccine Technology Roadmap illustrates that there is a political will among the global health community to make it happen.
Collapse
Affiliation(s)
- K L Wilson
- a Department of Immunology and Pathology, Faculty of Medicine, Nursing and Health Sciences , Monash University , Melbourne , Australia.,b School of Health and Biomedical Sciences , RMIT University , Bundoora , Australia
| | - K L Flanagan
- a Department of Immunology and Pathology, Faculty of Medicine, Nursing and Health Sciences , Monash University , Melbourne , Australia.,b School of Health and Biomedical Sciences , RMIT University , Bundoora , Australia.,c School of Medicine, Faculty of Health Sciences , University of Tasmania , Launceston , Australia
| | - M D Prakash
- b School of Health and Biomedical Sciences , RMIT University , Bundoora , Australia
| | - M Plebanski
- b School of Health and Biomedical Sciences , RMIT University , Bundoora , Australia
| |
Collapse
|
34
|
Kwenti TE, Kukwah TA, Kwenti TDB, Nyassa BR, Dilonga MH, Enow-Orock G, Tendongfor N, Anong ND, Wanji S, Njunda LA, Nkuo-Akenji T. Comparative analysis of IgG and IgG subclasses against Plasmodium falciparum MSP-1 19 in children from five contrasting bioecological zones of Cameroon. Malar J 2019; 18:16. [PMID: 30670064 PMCID: PMC6341684 DOI: 10.1186/s12936-019-2654-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 01/16/2019] [Indexed: 12/03/2022] Open
Abstract
Background Studies reporting the natural immune responses against malaria in children from different geographical settings in endemic areas are not readily available. This study was aimed at comparing the immune responses against Plasmodium falciparum MSP-119 antigen in children from five contrasting bioecological zones in Cameroon. Methods In a cross-sectional survey, children between 2 and 15 years, were enrolled from five ecological strata including the south Cameroonian equatorial forest, sudano-sahelian, high inland plateau, high western plateau, and the coastal strata. The children were screened for clinical malaria (defined by malaria parasitaemia ≥ 5000 parasites/µl plus axillary temperature ≥ 37.5 °C). Their antibody responses were measured against P. falciparum MSP-119 antigen using standard ELISA technique. Results In all, 415 children comprising 217 (52.3%) males participated. Total IgG and IgG1–IgG4 titres were observed to increase with age in all the strata except in the sudano-sahelian and high inland plateau strata. Total IgG and IgG1–IgG4 titres were significantly higher in the coastal strata and lowest in the high inland plateau (for IgG1 and IgG2) and sudano-sahelian strata (for IgG3 and IgG4). Titres of the cytophilic antibodies (IgG1 and IgG3) were significantly higher than the non-cytophilic antibodies (IgG2 and IgG4) in all the strata except in the sudano-sahelian and high inland plateau strata. Total IgG and IgG subclass titres were significantly higher in children positive for clinical malaria compared to negative children in all study sites except in the high western plateau and coastal (for IgG1 and IgG3), and the sudano-sahelian strata (for all antibodies). Furthermore, a significant positive correlation was observed between parasite density and IgG2 or IgG4 titres in all study sites except in the south Cameroonian equatorial forest and sudano-sahelian strata. Conclusions This study showed that antibody responses against MSP-119 vary considerably in children from the different bioecological strata in Cameroon and could be linked to the differential exposure to malaria in the different strata. Furthermore, the rate of antibody acquisition was not observed to increase in an age-dependent manner in low transmission settings. Electronic supplementary material The online version of this article (10.1186/s12936-019-2654-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tebit Emmanuel Kwenti
- Department of Medical Laboratory Sciences, Faculty of Health Science, University of Buea, P.O. Box 23, Buea, Southwest Region, Cameroon. .,Regional Hospital of Buea, P.O. Box 32, Buea, Southwest Region, Cameroon. .,Department of Public Health and Hygiene, Faculty of Health Science, University of Buea, P.O. Box 23, Buea, Southwest Region, Cameroon. .,Department of Microbiology and Parasitology, Faculty of Science, University of Buea, P.O. Box 63, Buea, Southwest Region, Cameroon.
| | - Tufon Anthony Kukwah
- Regional Hospital of Buea, P.O. Box 32, Buea, Southwest Region, Cameroon.,Department of Microbiology and Parasitology, Faculty of Science, University of Buea, P.O. Box 63, Buea, Southwest Region, Cameroon
| | - Tayong Dizzle Bita Kwenti
- Department of Microbiology and Parasitology, Faculty of Science, University of Buea, P.O. Box 63, Buea, Southwest Region, Cameroon
| | - Babila Raymond Nyassa
- Department of Microbiology and Parasitology, Faculty of Science, University of Buea, P.O. Box 63, Buea, Southwest Region, Cameroon
| | - Meriki Henry Dilonga
- Regional Hospital of Buea, P.O. Box 32, Buea, Southwest Region, Cameroon.,Department of Microbiology and Parasitology, Faculty of Science, University of Buea, P.O. Box 63, Buea, Southwest Region, Cameroon
| | - George Enow-Orock
- Regional Hospital of Buea, P.O. Box 32, Buea, Southwest Region, Cameroon.,Department of Biomedical Science, Faculty of Health Science, University of Buea, P.O Box 23, Buea, Southwest Region, Cameroon
| | - Nicholas Tendongfor
- Department of Public Health and Hygiene, Faculty of Health Science, University of Buea, P.O. Box 23, Buea, Southwest Region, Cameroon.,Department of Microbiology and Parasitology, Faculty of Science, University of Buea, P.O. Box 63, Buea, Southwest Region, Cameroon
| | - Nota Damian Anong
- Department of Biological Science, Faculty of Science, University of Bamenda, Bamenda, North West Region, Cameroon
| | - Samuel Wanji
- Parasites and Vector Biology Research Unit, Department of Microbiology and Parasitology, University of Buea, Buea, Cameroon
| | - Longdoh Anna Njunda
- Department of Medical Laboratory Sciences, Faculty of Health Science, University of Buea, P.O. Box 23, Buea, Southwest Region, Cameroon
| | - Theresa Nkuo-Akenji
- Department of Biological Science, Faculty of Science, University of Bamenda, Bamenda, North West Region, Cameroon
| |
Collapse
|
35
|
Abstract
The blood stage of the malaria parasite life cycle is responsible for all the clinical symptoms of malaria. During the blood stage, Plasmodium merozoites invade and multiply within host red blood cells (RBCs). Here, we review the progress made, challenges faced, and new strategies available for the development of blood stage malaria vaccines. We discuss our current understanding of immune responses against blood stages and the status of clinical development of various blood stage malaria vaccine candidates. We then discuss possible paths forward to develop effective blood stage malaria vaccines. This includes a discussion of protective immune mechanisms that can be elicited to target blood stage parasites, novel delivery systems, immunoassays and animal models to optimize vaccine candidates in preclinical studies, and use of challenge models to get an early readout of vaccine efficacy.
Collapse
|
36
|
Silveira ELV, Dominguez MR, Soares IS. To B or Not to B: Understanding B Cell Responses in the Development of Malaria Infection. Front Immunol 2018; 9:2961. [PMID: 30619319 PMCID: PMC6302011 DOI: 10.3389/fimmu.2018.02961] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 12/03/2018] [Indexed: 12/18/2022] Open
Abstract
Malaria is a widespread disease caused mainly by the Plasmodium falciparum (Pf) and Plasmodium vivax (Pv) protozoan parasites. Depending on the parasite responsible for the infection, high morbidity and mortality can be triggered. To escape the host immune responses, Plasmodium parasites disturb the functionality of B cell subsets among other cell types. However, some antibodies elicited during a malaria infection have the potential to block pathogen invasion and dissemination into the host. Thus, the question remains, why is protection not developed and maintained after the primary parasite exposure? In this review, we discuss different aspects of B cell responses against Plasmodium antigens during malaria infection. Since most studies have focused on the quantification of serum antibody titers, those B cell responses have not been fully characterized. However, to secrete antibodies, a complex cellular response is set up, including not only the activation and differentiation of B cells into antibody-secreting cells, but also the participation of other cell subsets in the germinal center reactions. Therefore, a better understanding of how B cell subsets are stimulated during malaria infection will provide essential insights toward the design of potent interventions.
Collapse
Affiliation(s)
- Eduardo L V Silveira
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Mariana R Dominguez
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Irene S Soares
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
37
|
Kamuyu G, Tuju J, Kimathi R, Mwai K, Mburu J, Kibinge N, Chong Kwan M, Hawkings S, Yaa R, Chepsat E, Njunge JM, Chege T, Guleid F, Rosenkranz M, Kariuki CK, Frank R, Kinyanjui SM, Murungi LM, Bejon P, Färnert A, Tetteh KKA, Beeson JG, Conway DJ, Marsh K, Rayner JC, Osier FHA. KILchip v1.0: A Novel Plasmodium falciparum Merozoite Protein Microarray to Facilitate Malaria Vaccine Candidate Prioritization. Front Immunol 2018; 9:2866. [PMID: 30619257 PMCID: PMC6298441 DOI: 10.3389/fimmu.2018.02866] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 11/21/2018] [Indexed: 12/12/2022] Open
Abstract
Passive transfer studies in humans clearly demonstrated the protective role of IgG antibodies against malaria. Identifying the precise parasite antigens that mediate immunity is essential for vaccine design, but has proved difficult. Completion of the Plasmodium falciparum genome revealed thousands of potential vaccine candidates, but a significant bottleneck remains in their validation and prioritization for further evaluation in clinical trials. Focusing initially on the Plasmodium falciparum merozoite proteome, we used peer-reviewed publications, multiple proteomic and bioinformatic approaches, to select and prioritize potential immune targets. We expressed 109 P. falciparum recombinant proteins, the majority of which were obtained using a mammalian expression system that has been shown to produce biologically functional extracellular proteins, and used them to create KILchip v1.0: a novel protein microarray to facilitate high-throughput multiplexed antibody detection from individual samples. The microarray assay was highly specific; antibodies against P. falciparum proteins were detected exclusively in sera from malaria-exposed but not malaria-naïve individuals. The intensity of antibody reactivity varied as expected from strong to weak across well-studied antigens such as AMA1 and RH5 (Kruskal–Wallis H test for trend: p < 0.0001). The inter-assay and intra-assay variability was minimal, with reproducible results obtained in re-assays using the same chip over a duration of 3 months. Antibodies quantified using the multiplexed format in KILchip v1.0 were highly correlated with those measured in the gold-standard monoplex ELISA [median (range) Spearman's R of 0.84 (0.65–0.95)]. KILchip v1.0 is a robust, scalable and adaptable protein microarray that has broad applicability to studies of naturally acquired immunity against malaria by providing a standardized tool for the detection of antibody correlates of protection. It will facilitate rapid high-throughput validation and prioritization of potential Plasmodium falciparum merozoite-stage antigens paving the way for urgently needed clinical trials for the next generation of malaria vaccines.
Collapse
Affiliation(s)
- Gathoni Kamuyu
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya.,Centre for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - James Tuju
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya.,Department of Biochemistry, Pwani University, Kilifi, Kenya
| | - Rinter Kimathi
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya
| | - Kennedy Mwai
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya
| | - James Mburu
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya
| | - Nelson Kibinge
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya
| | - Marisa Chong Kwan
- Arrayjet, Innovative Microarray Solutions, Edinburgh, United Kingdom
| | - Sam Hawkings
- Arrayjet, Innovative Microarray Solutions, Edinburgh, United Kingdom
| | - Reuben Yaa
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya
| | - Emily Chepsat
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya
| | - James M Njunge
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya
| | - Timothy Chege
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya
| | - Fatuma Guleid
- Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Micha Rosenkranz
- Centre for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Christopher K Kariuki
- Department of Tropical and Infectious Diseases, Institute of Primate Research, Nairobi, Kenya.,Cellular and Molecular Immunology, Vrije Universiteit Brussels, Brussels, Belgium
| | - Roland Frank
- Centre for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Samson M Kinyanjui
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya.,Department of Biochemistry, Pwani University, Kilifi, Kenya.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Linda M Murungi
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya
| | - Philip Bejon
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya
| | - Anna Färnert
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Kevin K A Tetteh
- Immunology and Infection Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - James G Beeson
- Burnet Institute, Melbourne, VIC, Australia.,Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - David J Conway
- Pathogen Molecular Biology Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Kevin Marsh
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom.,African Academy of Sciences, Nairobi, Kenya
| | - Julian C Rayner
- Wellcome Sanger Institute, Hinxton, Cambridge, United Kingdom
| | - Faith H A Osier
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya.,Centre for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany.,Department of Biochemistry, Pwani University, Kilifi, Kenya.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
38
|
Amoah LE, Abagna HB, Akyea-Mensah K, Lo AC, Kusi KA, Gyan BA. Characterization of anti-EBA175RIII-V in asymptomatic adults and children living in communities in the Greater Accra Region of Ghana with varying malaria transmission intensities. BMC Immunol 2018; 19:34. [PMID: 30453898 PMCID: PMC6245760 DOI: 10.1186/s12865-018-0271-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 11/08/2018] [Indexed: 11/10/2022] Open
Abstract
Background Antibodies against Region III-V of the erythrocyte binding antigen (EBA) 175 (EBA175RIII-V) have been suggested to provide protection from malaria in a natural infection. However, the quality and quantity of naturally induced antibodies to EBA175RIII-V has not been fully characterized in different cohorts of Ghanaians. This study sought to determine the characteristics of antibodies against EBA175RIII-V in asymptomatic adults and children living in two communities of varying P. falciparum parasite prevalence in southern Ghana. Methods Microscopic evaluation of thick and thin blood smears was used to identify asymptomatic Plasmodium falciparum carriage and indirect enzyme linked immunosorbent (ELISA) used to assess antibody concentrations and avidity. Results Parasite carriage estimated by microscopy in Obom was 35.6% as opposed to 3.5% in Asutsuare. Levels of IgG, IgG1, IgG2, IgG3 and IgG4 against EBA175RIII-V in the participants from Obom were significantly higher (P < 0.05, Dunn’s Multiple Comparison test) than those in Asutsuare. However the relative avidity of IgG antibodies against EBA175RIII-V was significantly higher (P < 0.0001, Mann Whitney test) in Asutsuare than in Obom. Conclusions People living in communities with limited exposure to P. falciparum parasites have low quantities of high avidity antibodies against EBA175RIII-V whilst people living in communities with high exposure to the parasites have high quantities of age-dependent but low avidity antibodies against EBA175RIII-V. Electronic supplementary material The online version of this article (10.1186/s12865-018-0271-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- L E Amoah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana.
| | - H B Abagna
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - K Akyea-Mensah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - A C Lo
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana.,Present address: University Cheikh Anta DIOP, Dakar, Senegal
| | - K A Kusi
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - B A Gyan
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| |
Collapse
|
39
|
Bittencourt NC, Leite JA, Silva ABIE, Pimenta TS, Silva-Filho JL, Cassiano GC, Lopes SCP, Dos-Santos JCK, Bourgard C, Nakaya HI, da Silva Ventura AMR, Lacerda MVG, Ferreira MU, Machado RLD, Albrecht L, Costa FTM. Genetic sequence characterization and naturally acquired immune response to Plasmodium vivax Rhoptry Neck Protein 2 (PvRON2). Malar J 2018; 17:401. [PMID: 30382855 PMCID: PMC6208078 DOI: 10.1186/s12936-018-2543-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 10/22/2018] [Indexed: 12/28/2022] Open
Abstract
Background The genetic diversity of malaria antigens often results in allele variant-specific immunity, imposing a great challenge to vaccine development. Rhoptry Neck Protein 2 (PvRON2) is a blood-stage antigen that plays a key role during the erythrocyte invasion of Plasmodium vivax. This study investigates the genetic diversity of PvRON2 and the naturally acquired immune response to P. vivax isolates. Results Here, the genetic diversity of PvRON21828–2080 and the naturally acquired humoral immune response against PvRON21828–2080 in infected and non-infected individuals from a vivax malaria endemic area in Brazil was reported. The diversity analysis of PvRON21828–2080 revealed that the protein is conserved in isolates in Brazil and worldwide. A total of 18 (19%) patients had IgG antibodies to PvRON21828–2080. Additionally, the analysis of the antibody response in individuals who were not acutely infected with malaria, but had been infected with malaria in the past indicated that 32 patients (33%) exhibited an IgG immune response against PvRON2. Conclusions PvRON2 was conserved among the studied isolates. The presence of naturally acquired antibodies to this protein in the absence of the disease suggests that PvRON2 induces a long-term antibody response. These results indicate that PvRON2 is a potential malaria vaccine candidate. Electronic supplementary material The online version of this article (10.1186/s12936-018-2543-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Najara C Bittencourt
- Laboratory of Tropical Diseases-Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Juliana A Leite
- Laboratory of Tropical Diseases-Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | | | - Tamirys S Pimenta
- Laboratório de Ensaios Clínicos e Imunogenética em Malária, Instituto Evandro Chagas/SVS/MS, Ananindeua, PA, Brazil
| | - João Luiz Silva-Filho
- Laboratory of Tropical Diseases-Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Gustavo C Cassiano
- Laboratory of Tropical Diseases-Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Stefanie C P Lopes
- Instituto Leônidas & Maria Deane, Fundação Oswaldo Cruz - FIOCRUZ, Manaus, AM, Brazil.,Fundação de Medicina Tropical-Dr. Heitor Vieira Dourado, Manaus, AM, Brazil
| | - Joao C K Dos-Santos
- Laboratory of Tropical Diseases-Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Catarina Bourgard
- Laboratory of Tropical Diseases-Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Helder I Nakaya
- School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Marcus V G Lacerda
- Instituto Leônidas & Maria Deane, Fundação Oswaldo Cruz - FIOCRUZ, Manaus, AM, Brazil.,Fundação de Medicina Tropical-Dr. Heitor Vieira Dourado, Manaus, AM, Brazil
| | - Marcelo U Ferreira
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo-USP, São Paulo, Brazil
| | - Ricardo L D Machado
- Laboratório de Ensaios Clínicos e Imunogenética em Malária, Instituto Evandro Chagas/SVS/MS, Ananindeua, PA, Brazil
| | - Letusa Albrecht
- Instituto Carlos Chagas, Fundação Oswaldo Cruz - FIOCRUZ, Curitiba, PR, Brazil.
| | - Fabio T M Costa
- Laboratory of Tropical Diseases-Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas-UNICAMP, Campinas, SP, Brazil.
| |
Collapse
|
40
|
Saavedra-Langer R, Marapara J, Valle-Campos A, Durand S, Vásquez-Chasnamote ME, Silva H, Pinedo-Cancino V. IgG subclass responses to excreted-secreted antigens of Plasmodium falciparum in a low-transmission malaria area of the Peruvian Amazon. Malar J 2018; 17:328. [PMID: 30200987 PMCID: PMC6131892 DOI: 10.1186/s12936-018-2471-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 08/30/2018] [Indexed: 11/10/2022] Open
Abstract
Background Malaria in Peru is concentrated in the Amazon region, especially in Loreto, and transmission is focused in rural and peri-urban communities. The government has approved a malaria elimination plan with a community approach and seeks to reduce the risk of transmission through preventive interventions, but asymptomatic and low-parasite-density infections are challenges for disease control and elimination. IgG antibodies play a critical role in combating infection through their ability to reduce parasitaemia and clinical symptoms. In particular, IgG subclasses have important roles in controlling malaria disease and may provide new insight into the development of malaria control strategies and understanding of malaria transmission. Through the use of excreted-secreted antigens from Plasmodium falciparum, were evaluated the responses of the four IgG subclasses in symptomatic and asymptomatic malarial infections. Results Higher levels of whole IgG were observed in asymptomatic carriers (P < 0.05). IgG3 and IgG1 were the most prevalent subclasses and did not show differences in their antibody levels in either type of carrier. All symptomatic carriers were positive for IgG4, and the presence of IgG3 and IgG2 were correlated with protection against parasitaemia. IgG2 showed lower prevalence and antibody titers in comparison to other subclasses. Conclusions This is the first study that characterizes the IgG subclass response in the Peruvian Amazon, and these results show that even in populations from regions with low malaria transmission, a certain degree of naturally acquired immunity can develop when the right antibody subclasses are produced. This provides important insight into the potential mechanisms regulating protective immunity. Electronic supplementary material The online version of this article (10.1186/s12936-018-2471-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rafael Saavedra-Langer
- Fundación para el Desarrollo Sostenible de la Amazonía Baja del Perú, Universidad Nacional de la Amazonía Peruana, Iquitos, Perú.,Laboratorio de Investigación de Productos Naturales Antiparasitarios de la Amazonía Peruana, Centro de Investigación de Recursos Naturales de la Amazonía, Universidad Nacional de la Amazonía Peruana, Iquitos, Perú
| | - Jorge Marapara
- Laboratorio de Investigación de Productos Naturales Antiparasitarios de la Amazonía Peruana, Centro de Investigación de Recursos Naturales de la Amazonía, Universidad Nacional de la Amazonía Peruana, Iquitos, Perú
| | - Andree Valle-Campos
- Fundación para el Desarrollo Sostenible de la Amazonía Baja del Perú, Universidad Nacional de la Amazonía Peruana, Iquitos, Perú
| | - Salomón Durand
- Centro de Investigación en Enfermedades Tropicales "Maxime Kuczynski", Instituto Nacional de Salud, Lima, Perú
| | - Maria E Vásquez-Chasnamote
- Fundación para el Desarrollo Sostenible de la Amazonía Baja del Perú, Universidad Nacional de la Amazonía Peruana, Iquitos, Perú.,Laboratorio de Investigación de Productos Naturales Antiparasitarios de la Amazonía Peruana, Centro de Investigación de Recursos Naturales de la Amazonía, Universidad Nacional de la Amazonía Peruana, Iquitos, Perú
| | - Hermann Silva
- Facultad de Medicina Humana, Universidad Nacional de la Amazonía Peruana, Iquitos, Perú
| | - Viviana Pinedo-Cancino
- Fundación para el Desarrollo Sostenible de la Amazonía Baja del Perú, Universidad Nacional de la Amazonía Peruana, Iquitos, Perú. .,Laboratorio de Investigación de Productos Naturales Antiparasitarios de la Amazonía Peruana, Centro de Investigación de Recursos Naturales de la Amazonía, Universidad Nacional de la Amazonía Peruana, Iquitos, Perú.
| |
Collapse
|
41
|
Kinetics of antibody responses to PfRH5-complex antigens in Ghanaian children with Plasmodium falciparum malaria. PLoS One 2018; 13:e0198371. [PMID: 29883485 PMCID: PMC5993283 DOI: 10.1371/journal.pone.0198371] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/17/2018] [Indexed: 11/21/2022] Open
Abstract
Plasmodium falciparum PfRH5 protein binds Ripr, CyRPA and Pf113 to form a complex that is essential for merozoite invasion of erythrocytes. The inter-genomic conservation of the PfRH5 complex proteins makes them attractive blood stage vaccine candidates. However, little is known about how antibodies to PfRH5, CyRPA and Pf113 are acquired and maintained in naturally exposed populations, and the role of PfRH5 complex proteins in naturally acquired immunity. To provide such data, we studied 206 Ghanaian children between the ages of 1–12 years, who were symptomatic, asymptomatic or aparasitemic and healthy. Plasma levels of antigen-specific IgG and IgG subclasses were measured by ELISA at several time points during acute disease and convalescence. On the day of admission with acute P. falciparum malaria, the prevalence of antibodies to PfRH5-complex proteins was low compared to other merozoite antigens (EBA175, GLURP-R0 and GLURP-R2). At convalescence, the levels of RH5-complex-specific IgG were reduced, with the decay of PfRH5-specific IgG being slower than the decay of IgG specific for CyRPA and Pf113. No correlation between IgG levels and protection against P. falciparum malaria was observed for any of the PfRH5 complex proteins. From this we conclude that specific IgG was induced against proteins from the PfRH5-complex during acute P. falciparum malaria, but the prevalence was low and the IgG levels decayed rapidly after treatment. These data indicate that the levels of IgG specific for PfRH5-complex proteins in natural infections in Ghanaian children were markers of recent exposure only.
Collapse
|
42
|
Ubillos I, Jiménez A, Vidal M, Bowyer PW, Gaur D, Dutta S, Gamain B, Coppel R, Chauhan V, Lanar D, Chitnis C, Angov E, Beeson J, Cavanagh D, Campo JJ, Aguilar R, Dobaño C. Optimization of incubation conditions of Plasmodium falciparum antibody multiplex assays to measure IgG, IgG 1-4, IgM and IgE using standard and customized reference pools for sero-epidemiological and vaccine studies. Malar J 2018; 17:219. [PMID: 29859096 PMCID: PMC5984756 DOI: 10.1186/s12936-018-2369-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/28/2018] [Indexed: 12/15/2022] Open
Abstract
Background The quantitative suspension array technology (qSAT) is a useful platform for malaria immune marker discovery. However, a major challenge for large sero-epidemiological and malaria vaccine studies is the comparability across laboratories, which requires the access to standardized control reagents for assay optimization, to monitor performance and improve reproducibility. Here, the Plasmodium falciparum antibody reactivities of the newly available WHO reference reagent for anti-malaria human plasma (10/198) and of additional customized positive controls were examined with seven in-house qSAT multiplex assays measuring IgG, IgG1–4 subclasses, IgM and IgE against a panel of 40 antigens. The different positive controls were tested at different incubation times and temperatures (4 °C overnight, 37 °C 2 h, room temperature 1 h) to select the optimal conditions. Results Overall, the WHO reference reagent had low IgG2, IgG4, IgM and IgE, and also low anti-CSP antibody levels, thus this reagent was enriched with plasmas from RTS,S-vaccinated volunteers to be used as standard for CSP-based vaccine studies. For the IgM assay, another customized plasma pool prepared with samples from malaria primo-infected adults with adequate IgM levels proved to be more adequate as a positive control. The range and magnitude of IgG and IgG1–4 responses were highest when the WHO reference reagent was incubated with antigen-coupled beads at 4 °C overnight. IgG levels measured in the negative control did not vary between incubations at 37 °C 2 h and 4 °C overnight, indicating no difference in unspecific binding. Conclusions With this study, the immunogenicity profile of the WHO reference reagent, including seven immunoglobulin isotypes and subclasses, and more P. falciparum antigens, also those included in the leading RTS,S malaria vaccine, was better characterized. Overall, incubation of samples at 4 °C overnight rendered the best performance for antibody measurements against the antigens tested. Although the WHO reference reagent performed well to measure IgG to the majority of the common P. falciparum blood stage antigens tested, customized pools may need to be used as positive controls depending on the antigens (e.g. pre-erythrocytic proteins of low natural immunogenicity) and isotypes/subclasses (e.g. IgM) under study. Electronic supplementary material The online version of this article (10.1186/s12936-018-2369-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Itziar Ubillos
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Carrer Rosselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain
| | - Alfons Jiménez
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Carrer Rosselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain.,CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Marta Vidal
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Carrer Rosselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain
| | - Paul W Bowyer
- Bacteriology Division, MHRA-NIBSC, South Mimms, Potter Bars, EN6 3QG, UK
| | - Deepak Gaur
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.,Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Sheetij Dutta
- U.S. Military Malaria Vaccine Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Benoit Gamain
- Université Sorbonne Paris Cité, Université Paris Diderot, Inserm, INTS, Unité Biologie Intégrée du Globule Rouge UMR_S1134, Laboratoire d'Excellence GR-Ex, Paris, France
| | - Ross Coppel
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Virander Chauhan
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - David Lanar
- U.S. Military Malaria Vaccine Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Chetan Chitnis
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Evelina Angov
- U.S. Military Malaria Vaccine Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - James Beeson
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, VIC, Australia
| | - David Cavanagh
- Institute of Immunology & Infection Research and Centre for Immunity, Infection & Evolution, Ashworth Laboratories, School of Biological Sciences, University of Edinburgh, King's Buildings, Charlotte Auerbach Rd, Edinburgh, EH9 3FL, UK
| | - Joseph J Campo
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Carrer Rosselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain
| | - Ruth Aguilar
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Carrer Rosselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain
| | - Carlota Dobaño
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Carrer Rosselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain.
| |
Collapse
|
43
|
Oyebola KM, Aina OO, Bah MM, Ajibaye S, Correa S, Awandare GA, Amambua-Ngwa A. Assessing naturally acquired immune response and malaria treatment outcomes in Lagos, Nigeria. AAS Open Res 2018. [DOI: 10.12688/aasopenres.12828.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background: There are emerging reports of poor efficacy of artemisinin-based combination treatment (ACT). However, mutations on the Kelch-13 gene marking delayed parasite clearance have no clinically defined relationship with ACT resistance across Africa. With increasing malaria control efforts, declining acquired immunity could be responsible for varying drug response profiles that may be dependent on levels of exposure to infections. To examine antibody responses against malaria and the influence on the efficacy of artemether-lumefantrine (AL), plasma samples were collected, prior to treatment, from individuals presenting with uncomplicated malaria. Methods: Participants were stratified into two groups: early (within 24 hours, N = 20) and late (between 48 – 72 hours, N = 30) parasite clearance after treatment, as determined by var gene acidic terminal sequence (varATS) polymerase chain reaction. Magnetic bead-based luminex assay was used to profile antibody responses specific to a panel of 21 Plasmodium falciparum sporozoite, merozoite and An. gambiae salivary antigens. Results: Median fluorescence intensity (MFI) of the antibodies was highest against glutamate-rich protein (GLURP-R0) and lowest against merozoite surface protein (MSP2) antigen. Analysis showed a positive correlation between expression of immunity and age of individuals (P = 0.023). However, there was no association between parasite density and antibody responses, except a significant positive relationship with reticulocyte binding protein-like homologue 5 (Rh5), P = 0.047; Plasmodium exported protein (Hyp2), P = 0.037 and merozoite surface protein 11 (H103), P = 0.038. Though higher levels of antibodies against erythrocyte binding antigens (EBA 140 and 175), MSP1.19, GLURP, circumsporozoite protein (CSP) and Rh4.2 were observed in individuals who recorded early parasite clearance, there was no significant difference in antibody responses in the early and late parasitological response groups. Conclusions: Characterization of additional markers in larger populations is required to reveal potential immunological correlates of drug efficacy.
Collapse
|
44
|
Abagna HB, Acquah FK, Okonu R, Aryee NA, Theisen M, Amoah LE. Assessment of the quality and quantity of naturally induced antibody responses to EBA175RIII-V in Ghanaian children living in two communities with varying malaria transmission patterns. Malar J 2018; 17:14. [PMID: 29310662 PMCID: PMC5759240 DOI: 10.1186/s12936-017-2167-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/29/2017] [Indexed: 01/26/2023] Open
Abstract
Background Recent global reports on malaria suggest significant decrease in disease severity and an increase in control interventions in many malaria endemic countries, including Ghana. However, a major driving force sustaining malaria transmission in recent times is the asymptomatic carriage of malaria parasites, which can enhance immune responses against parasite antigens. This study determined the prevalence and relative avidities of naturally induced antibodies to EBA175RIII–VLl in asymptomatic children living in two communities with varying malaria transmission patterns. Methods An asexual stage Plasmodium falciparum antigen, EBA175RIII–VLl was expressed in Lactococcus lactis, purified and used in indirect ELISA to measure total and cytophilic IgG concentrations and avidities in children aged between 6 and 12 years. The children were selected from Obom and Abura, communities with perennial and seasonal malaria transmission, respectively. Venous blood samples were collected in July and October 2015 and again in January 2016. The multiplicity of infection and the genetic diversity of EBA175RIII circulating in both sites were also assessed using polymerase chain reaction. Results Asymptomatic parasite carriage in the children from Obom decreased from July (peak season), through October and January, however parasite carriage in children from Abura was bimodal, with the lowest prevalence estimated in October. Antibody concentrations over the course of the study remained stable within each study site however, children living in Obom had significantly higher EBA175RIII–VLl antibody concentrations than children living in Abura (P < 0.05, Mann–Whitney test). Over the course of the study, the relative antibody avidities of EBA175RIII–VLl IgG antibodies were similar within and between the sites. Conclusion Naturally acquired IgG concentrations but not relative antibody avidities to EBA175RIII–V were significantly higher in Obom where malaria transmission is perennial than in Abura, where malaria transmission is seasonal. Electronic supplementary material The online version of this article (10.1186/s12936-017-2167-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hamza B Abagna
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana.,Department of Medical Biochemistry, University of Ghana, Accra, Ghana
| | - Festus K Acquah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Ruth Okonu
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Nii A Aryee
- Department of Medical Biochemistry, University of Ghana, Accra, Ghana
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Linda E Amoah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana.
| |
Collapse
|
45
|
Payne RO, Silk SE, Elias SC, Miura K, Diouf A, Galaway F, de Graaf H, Brendish NJ, Poulton ID, Griffiths OJ, Edwards NJ, Jin J, Labbé GM, Alanine DG, Siani L, Di Marco S, Roberts R, Green N, Berrie E, Ishizuka AS, Nielsen CM, Bardelli M, Partey FD, Ofori MF, Barfod L, Wambua J, Murungi LM, Osier FH, Biswas S, McCarthy JS, Minassian AM, Ashfield R, Viebig NK, Nugent FL, Douglas AD, Vekemans J, Wright GJ, Faust SN, Hill AV, Long CA, Lawrie AM, Draper SJ. Human vaccination against RH5 induces neutralizing antimalarial antibodies that inhibit RH5 invasion complex interactions. JCI Insight 2017; 2:96381. [PMID: 29093263 PMCID: PMC5752323 DOI: 10.1172/jci.insight.96381] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/05/2017] [Indexed: 11/17/2022] Open
Abstract
The development of a highly effective vaccine remains a key strategic goal to aid the control and eventual eradication of Plasmodium falciparum malaria. In recent years, the reticulocyte-binding protein homolog 5 (RH5) has emerged as the most promising blood-stage P. falciparum candidate antigen to date, capable of conferring protection against stringent challenge in Aotus monkeys. We report on the first clinical trial to our knowledge to assess the RH5 antigen - a dose-escalation phase Ia study in 24 healthy, malaria-naive adult volunteers. We utilized established viral vectors, the replication-deficient chimpanzee adenovirus serotype 63 (ChAd63), and the attenuated orthopoxvirus modified vaccinia virus Ankara (MVA), encoding RH5 from the 3D7 clone of P. falciparum. Vaccines were administered i.m. in a heterologous prime-boost regimen using an 8-week interval and were well tolerated. Vaccine-induced anti-RH5 serum antibodies exhibited cross-strain functional growth inhibition activity (GIA) in vitro, targeted linear and conformational epitopes within RH5, and inhibited key interactions within the RH5 invasion complex. This is the first time to our knowledge that substantial RH5-specific responses have been induced by immunization in humans, with levels greatly exceeding the serum antibody responses observed in African adults following years of natural malaria exposure. These data support the progression of RH5-based vaccines to human efficacy testing.
Collapse
Affiliation(s)
- Ruth O. Payne
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Sarah E. Silk
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Sean C. Elias
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, Maryland, USA
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, Maryland, USA
| | - Francis Galaway
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Hans de Graaf
- NIHR Wellcome Trust Clinical Research Facility, University Hospital Southampton NHS Foundation Trust and Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Nathan J. Brendish
- NIHR Wellcome Trust Clinical Research Facility, University Hospital Southampton NHS Foundation Trust and Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Ian D. Poulton
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Nick J. Edwards
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Jing Jin
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | | - Loredana Siani
- ReiThera SRL (formerly Okairos SRL), Viale Città d’Europa, Rome, Italy
| | - Stefania Di Marco
- ReiThera SRL (formerly Okairos SRL), Viale Città d’Europa, Rome, Italy
| | - Rachel Roberts
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Nicky Green
- Clinical Biomanufacturing Facility, University of Oxford, Oxford, United Kingdom
| | - Eleanor Berrie
- Clinical Biomanufacturing Facility, University of Oxford, Oxford, United Kingdom
| | | | | | - Martino Bardelli
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Frederica D. Partey
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
- Centre for Medical Parasitology, Department of Immunology and Microbiology (ISIM), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Michael F. Ofori
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Lea Barfod
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Juliana Wambua
- KEMRI Centre for Geographic Medicine Research, Kilifi, Kenya
| | - Linda M. Murungi
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
- KEMRI Centre for Geographic Medicine Research, Kilifi, Kenya
| | - Faith H. Osier
- KEMRI Centre for Geographic Medicine Research, Kilifi, Kenya
| | - Sumi Biswas
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - James S. McCarthy
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | | | - Rebecca Ashfield
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Nicola K. Viebig
- European Vaccine Initiative, UniversitätsKlinikum Heidelberg, Heidelberg, Germany
| | - Fay L. Nugent
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | | - Gavin J. Wright
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Saul N. Faust
- NIHR Wellcome Trust Clinical Research Facility, University Hospital Southampton NHS Foundation Trust and Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Adrian V.S. Hill
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Carole A. Long
- Laboratory of Malaria and Vector Research, NIAID/NIH, Rockville, Maryland, USA
| | - Alison M. Lawrie
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Simon J. Draper
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
46
|
Valmaseda A, Bassat Q, Aide P, Cisteró P, Jiménez A, Casellas A, Machevo S, Aguilar R, Sigaúque B, Chauhan VS, Langer C, Beeson J, Chitnis C, Alonso PL, Gaur D, Mayor A. Host age and expression of genes involved in red blood cell invasion in Plasmodium falciparum field isolates. Sci Rep 2017; 7:4717. [PMID: 28680086 PMCID: PMC5498679 DOI: 10.1038/s41598-017-05025-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 05/23/2017] [Indexed: 11/24/2022] Open
Abstract
Plasmodium falciparum proteins involved in erythrocyte invasion are main targets of acquired immunity and important vaccine candidates. We hypothesized that anti-parasite immunity acquired upon exposure would limit invasion-related gene (IRG) expression and affect the clinical impact of the infection. 11 IRG transcript levels were measured in P. falciparum isolates by RT-PCR, and IgG/IgM against invasion ligands by Luminex®, in 50 Mozambican adults, 25 children with severe malaria (SM) and 25 with uncomplicated malaria (UM). IRG expression differences among groups and associations between IRG expression and clinical/immunologic parameters were assessed. IRG expression diversity was higher in parasites infecting children than adults (p = 0.022). eba140 and ptramp expression decreased with age (p = 0.003 and 0.007, respectively) whereas p41 expression increased (p = 0.022). pfrh5 reduction in expression was abrupt early in life. Parasite density decreased with increasing pfrh5 expression (p < 0.001) and, only in children, parasite density increased with p41 expression (p = 0.007), and decreased with eba175 (p = 0.013). Antibody responses and IRG expression were not associated. In conclusion, IRG expression is associated with age and parasite density, but not with specific antibody responses in the acute phase of infection. Our results confirm the importance of multi-antigen vaccines development to avoid parasite immune escape when tested in malaria-exposed individuals.
Collapse
Affiliation(s)
- Aida Valmaseda
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.
| | - Quique Bassat
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde de Manhiça (CISM), Manhiça, Mozambique.,ICREA, Pg. Lluís Companys 23, 08010, Barcelona, Spain
| | - Pedro Aide
- Centro de Investigação em Saúde de Manhiça (CISM), Manhiça, Mozambique
| | - Pau Cisteró
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Alfons Jiménez
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBEREsp), Madrid, Spain
| | - Aina Casellas
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Sonia Machevo
- Centro de Investigação em Saúde de Manhiça (CISM), Manhiça, Mozambique
| | - Ruth Aguilar
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Betuel Sigaúque
- Centro de Investigação em Saúde de Manhiça (CISM), Manhiça, Mozambique
| | - Virander S Chauhan
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Christine Langer
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
| | - James Beeson
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
| | - Chetan Chitnis
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Pedro L Alonso
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde de Manhiça (CISM), Manhiça, Mozambique
| | - Deepak Gaur
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Alfredo Mayor
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain. .,Centro de Investigação em Saúde de Manhiça (CISM), Manhiça, Mozambique.
| |
Collapse
|
47
|
Acharya P, Garg M, Kumar P, Munjal A, Raja KD. Host-Parasite Interactions in Human Malaria: Clinical Implications of Basic Research. Front Microbiol 2017; 8:889. [PMID: 28572796 PMCID: PMC5435807 DOI: 10.3389/fmicb.2017.00889] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 05/02/2017] [Indexed: 12/21/2022] Open
Abstract
The malaria parasite, Plasmodium, is one of the oldest parasites documented to infect humans and has proven particularly hard to eradicate. One of the major hurdles in designing an effective subunit vaccine against the malaria parasite is the insufficient understanding of host–parasite interactions within the human host during infections. The success of the parasite lies in its ability to evade the human immune system and recruit host responses as physiological cues to regulate its life cycle, leading to rapid acclimatization of the parasite to its immediate host environment. Hence understanding the environmental niche of the parasite is crucial in developing strategies to combat this deadly infectious disease. It has been increasingly recognized that interactions between parasite proteins and host factors are essential to establishing infection and virulence at every stage of the parasite life cycle. This review reassesses all of these interactions and discusses their clinical importance in designing therapeutic approaches such as design of novel vaccines. The interactions have been followed from the initial stages of introduction of the parasite under the human dermis until asexual and sexual blood stages which are essential for transmission of malaria. We further classify the interactions as “direct” or “indirect” depending upon their demonstrated ability to mediate direct physical interactions of the parasite with host factors or their indirect manipulation of the host immune system since both forms of interactions are known to have a crucial role during infections. We also discuss the many ways in which this understanding has been taken to the field and the success of these strategies in controlling human malaria.
Collapse
Affiliation(s)
- Pragyan Acharya
- Department of Biochemistry, All India Institute of Medical SciencesNew Delhi, India
| | - Manika Garg
- Department of Biochemistry, Jamia Hamdard UniversityNew Delhi, India
| | - Praveen Kumar
- Department of Biochemistry, All India Institute of Medical SciencesNew Delhi, India
| | - Akshay Munjal
- Department of Biochemistry, All India Institute of Medical SciencesNew Delhi, India
| | - K D Raja
- Department of Biochemistry, All India Institute of Medical SciencesNew Delhi, India
| |
Collapse
|