1
|
Kisalu NK, Silva Pereira LD, Herman JD, Asokan M, Ernste K, Merriam J, Liu C, DeMouth ME, Pegu A, Lofgren M, Dillon M, Bonilla B, MacVicar R, Zur Y, Kiyuka P, Flores-Garcia Y, Chakraborty S, Nikolaeva D, Ogwang R, Flynn B, Francica J, Pierson TC, Koup RA, Zavala F, Wang TT, Alter G, Idris AH, Seder RA. FcγR binding differentially contributes to protection by two human monoclonal antibodies targeting Plasmodium falciparum circumsporozoite protein. Sci Transl Med 2025; 17:eadk6745. [PMID: 40267218 DOI: 10.1126/scitranslmed.adk6745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 09/25/2024] [Accepted: 03/05/2025] [Indexed: 04/25/2025]
Abstract
Antibodies mediate protection against a wide range of pathogens through binding and neutralizing the pathogen or through Fc-mediated effector functions. Human monoclonal antibodies (mAbs) CIS43LS and L9LS show high-affinity binding targeting distinct regions on the Plasmodium falciparum circumsporozoite protein (PfCSP) and are highly effective in preventing malaria in humans. However, the role of FcγR binding in protection by these mAbs has not been determined. Here, we assessed several Fc variants of CIS43LS and L9LS for protection against infection with transgenic Plasmodium berghei parasite expressing PfCSP in mice. Limiting binding to FcγRs did not reduce protection compared to the parental mAbs in mice. To determine whether protection could be improved in vivo by Fc modification, we engineered Fc variant mAbs with increased binding to distinct FcγRs. Passive transfer of CIS43LS-DE and CIS43LS-DEAL variants resulted in an approximately two- to threefold reduction in the liver-stage parasite burden in C57BL/6 or human FcγR mice compared with the parental CIS43LS after challenge. CIS43LS-DEAL also enhanced protection of mice after mosquito bite challenge. Systems serology analysis revealed that the CIS43LS-DE and CIS43LS-DEAL variants could enhance human neutrophil and monocyte phagocytosis, as well as NK cell activation, compared with CIS43LS. However, similar Fc modifications incorporated into L9LS did not increase protection compared to the parental mAb. Overall, although FcγR binding by CIS43LS and L9LS is dispensable in mouse models of malaria, enhancing the binding of CIS43LS to FcγR showed a modest increase in the potency of this mAb.
Collapse
Affiliation(s)
- Neville K Kisalu
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Lais D Silva Pereira
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Jonathan D Herman
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Mangaiarkarasi Asokan
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Keenan Ernste
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Jonah Merriam
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Cuiping Liu
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Megan E DeMouth
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Amarendra Pegu
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Mariah Lofgren
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Marlon Dillon
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Brian Bonilla
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Ryan MacVicar
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Yonatan Zur
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Patience Kiyuka
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Yevel Flores-Garcia
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Saborni Chakraborty
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine and Department of Medicine, Division of Infectious Diseases, Stanford University, Stanford, CA 94305, USA
| | - Daria Nikolaeva
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rodney Ogwang
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Barbara Flynn
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Joseph Francica
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Theodore C Pierson
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Richard A Koup
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | - Fidel Zavala
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Taia T Wang
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine and Department of Medicine, Division of Infectious Diseases, Stanford University, Stanford, CA 94305, USA
| | - Galit Alter
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Azza H Idris
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Robert A Seder
- Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
2
|
Boyle MJ, Engwerda CR, Jagannathan P. The impact of Plasmodium-driven immunoregulatory networks on immunity to malaria. Nat Rev Immunol 2024; 24:637-653. [PMID: 38862638 PMCID: PMC11688169 DOI: 10.1038/s41577-024-01041-5] [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] [Accepted: 04/30/2024] [Indexed: 06/13/2024]
Abstract
Malaria, caused by infection with Plasmodium parasites, drives multiple regulatory responses across the immune landscape. These regulatory responses help to protect against inflammatory disease but may in some situations hamper the acquisition of adaptive immune responses that clear parasites. In addition, the regulatory responses that occur during Plasmodium infection may negatively affect malaria vaccine efficacy in the most at-risk populations. Here, we discuss the specific cellular mechanisms of immunoregulatory networks that develop during malaria, with a focus on knowledge gained from human studies and studies that involve the main malaria parasite to affect humans, Plasmodium falciparum. Leveraging this knowledge may lead to the development of new therapeutic approaches to increase protective immunity to malaria during infection or after vaccination.
Collapse
Affiliation(s)
- Michelle J Boyle
- Life Sciences Division, Burnet Institute, Melbourne, Victoria, Australia.
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
| | | | - Prasanna Jagannathan
- Department of Medicine, Stanford University, Stanford, CA, USA.
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
3
|
Zhan Q, He Q, Tiedje KE, Day KP, Pascual M. Hyper-diverse antigenic variation and resilience to transmission-reducing intervention in falciparum malaria. Nat Commun 2024; 15:7343. [PMID: 39187488 PMCID: PMC11347654 DOI: 10.1038/s41467-024-51468-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 08/07/2024] [Indexed: 08/28/2024] Open
Abstract
Intervention efforts against falciparum malaria in high-transmission regions remain challenging, with rapid resurgence typically following their relaxation. Such resilience co-occurs with incomplete immunity and a large transmission reservoir from high asymptomatic prevalence. Incomplete immunity relates to the large antigenic variation of the parasite, with the major surface antigen of the blood stage of infection encoded by the multigene and recombinant family known as var. With a stochastic agent-based model, we investigate the existence of a sharp transition in resurgence ability with intervention intensity and identify molecular indicators informative of its proximity. Their application to survey data with deep sampling of var sequences from individual isolates in northern Ghana suggests that the transmission system was brought close to transition by intervention with indoor residual spraying. These results indicate that sustaining and intensifying intervention would have pushed malaria dynamics to a slow-rebound regime with an increased probability of local parasite extinction.
Collapse
Affiliation(s)
- Qi Zhan
- Committee on Genetics, Genomics and Systems Biology, The University of Chicago, Chicago, IL, 60637, USA
| | - Qixin He
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Kathryn E Tiedje
- Department of Microbiology and Immunology, Bio21 Institute and Peter Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - Karen P Day
- Department of Microbiology and Immunology, Bio21 Institute and Peter Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - Mercedes Pascual
- Department of Biology, New York University, New York, NY, 10003, USA.
- Department of Environmental Studies, New York University, New York, NY, 10003, USA.
- Santa Fe Institute, Santa Fe, NM, 87501, USA.
| |
Collapse
|
4
|
Ngu L, Fotso HO, Nyebe I, Tchadji JC, Ambada G, Ndah A, Atechi B, Lissom A, Atabonkeng PE, Chukwuma G, Efezeuh V, Gyu PC, Esimone C, Nguedia JCA, Akum EA, Okeke M, Titanji VPK, Mbacham W, Bopda-Waffo A, Wapimewah GN. Immunoglobulin G (IgG) specific responses to recombinant Qβ displayed MSP3 and UB05 in plasma of asymptomatic Plasmodium falciparum-infected children living in two different agro-ecological settings of Cameroon. Pan Afr Med J 2024; 47:175. [PMID: 39036016 PMCID: PMC11260061 DOI: 10.11604/pamj.2024.47.175.38169] [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: 11/10/2022] [Accepted: 02/25/2024] [Indexed: 07/23/2024] Open
Abstract
Introduction in areas with intense perennial malaria transmission, limited data is available on the impact of environmental conditions especially rainfall on naturally acquired immunity against promising malaria vaccine candidates. For this reason, we have compared IgG antibody responses specific to Plasmodium spp. derived MSP3 and UB05 vaccine candidates, in plasma of children living in two areas of Cameroon differing in rainfall conditions. Methods data about children less than 5 years old was collected during the years 2017 and 2018. Next malaria asymptomatic P. falciparum (Pf) infected children were selected following malaria test confirmation. MSP3 and UB05 specific IgG antibody responses were measured in participant´s plasma using enzyme-linked immunosorbent assay (ELISA). Results interestingly, IgG antibody responses specific to UB05 were significantly higher (p<0.0001) in Pf-negative children when compared to their asymptomatic Pf-infected counterparts living in monomodal rainfall areas. In contrast, a significantly higher (p<0.0001) IgG response to MSP3 was observed instead in asymptomatic Pf-infected children in the same population. In addition, IgG responses specific to UB05 remained significantly higher in bimodal when compared to monomodal rainfall areas irrespective of children´s Pf infection status (p<0.0055 for Pf-positive and p<0.0001 for negative children). On the contrary, IgG antibody responses specific to MSP3 were significantly higher in bimodal relative to monomodal rainfall areas (P<0.0001) just for Pf-negative children. Conclusion thus IgG antibody responses specific to UBO5 are a better correlate of naturally acquired immunity against malaria in Pf-negative Cameroonian children especially in monomodal rainfall areas.
Collapse
Affiliation(s)
- Loveline Ngu
- Laboratory of Vaccinology/Biobanking, Chantal Biya International Reference Center for Research on the Prevention and Management of HIV/AIDS, Yaounde, Cameroon
- Department of Biochemistry, Faculty of Sciences, University of Yaounde I, Yaounde, Cameroon
| | - Herve Ouambo Fotso
- Laboratory of Vaccinology/Biobanking, Chantal Biya International Reference Center for Research on the Prevention and Management of HIV/AIDS, Yaounde, Cameroon
- Pan African Center for Clinical and Translational Sciences (PANECTS), Yaounde, Cameroon
| | - Inès Nyebe
- Laboratory of Vaccinology/Biobanking, Chantal Biya International Reference Center for Research on the Prevention and Management of HIV/AIDS, Yaounde, Cameroon
- Pan African Center for Clinical and Translational Sciences (PANECTS), Yaounde, Cameroon
- Department of Microbiology, Faculty of Sciences, University of Yaounde I, Yaounde, Cameroon
| | - Jules Colince Tchadji
- Laboratory of Vaccinology/Biobanking, Chantal Biya International Reference Center for Research on the Prevention and Management of HIV/AIDS, Yaounde, Cameroon
- Department of Animal Biology and Physiology, Faculty Of Sciences, University of Yaounde I, Yaoundé, Cameroon
| | - Georgia Ambada
- Laboratory of Vaccinology/Biobanking, Chantal Biya International Reference Center for Research on the Prevention and Management of HIV/AIDS, Yaounde, Cameroon
- Department of Animal Biology and Physiology, Faculty Of Sciences, University of Yaounde I, Yaoundé, Cameroon
| | - Akeleke Ndah
- Pan African Center for Clinical and Translational Sciences (PANECTS), Yaounde, Cameroon
| | - Bloomfield Atechi
- Pan African Center for Clinical and Translational Sciences (PANECTS), Yaounde, Cameroon
| | - Abel Lissom
- Laboratory of Vaccinology/Biobanking, Chantal Biya International Reference Center for Research on the Prevention and Management of HIV/AIDS, Yaounde, Cameroon
- Department of Biological Sciences, Faculty of Sciences, University of Bamenda, Bamenda, Cameroon
| | | | - George Chukwuma
- Department of Medical Laboratory Science, College of Health Sciences, Nnamdi Azikiwe University, Awka, Nigeria
| | - Vitalis Efezeuh
- Department of Biochemistry and Molecular Biology, University of Buea, Buea, Cameroon
| | - Park Chae Gyu
- Laboratory of Immunology, Brain Korea 21 PLUS Project for Medical Science, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Charles Esimone
- Department of Pharmaceutical Microbiology and Biotechnology, Nnamdi Azikiwe University, Awka, Nigeria
| | | | - Eric Achidi Akum
- Department of Medical Laboratory Sciences, University of Buea, Buea, Cameroon
| | - Malachy Okeke
- Department of Natural and Environmental Sciences, Biomedical Science Concentration, School of Arts and Sciences, American University of Nigeria, 98 Lamido Zubairu Way, Yola, Nigeria
| | | | - Wilfred Mbacham
- Department of Biochemistry, Faculty of Sciences, University of Yaounde I, Yaounde, Cameroon
| | - Alain Bopda-Waffo
- Pan African Center for Clinical and Translational Sciences (PANECTS), Yaounde, Cameroon
- Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, MS1017Q Lab MS1015, Indianapolis, IN, United States of America
| | - Godwin Nchinda Wapimewah
- Laboratory of Vaccinology/Biobanking, Chantal Biya International Reference Center for Research on the Prevention and Management of HIV/AIDS, Yaounde, Cameroon
- Pan African Center for Clinical and Translational Sciences (PANECTS), Yaounde, Cameroon
- Department of Pharmaceutical Microbiology and Biotechnology, Nnamdi Azikiwe University, Awka, Nigeria
| |
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
|
Ofori EA, Garcia-Senosiain A, Naghizadeh M, Kana IH, Dziegiel MH, Adu B, Singh S, Theisen M. Human blood neutrophils generate ROS through FcγR-signaling to mediate protection against febrile P. falciparum malaria. Commun Biol 2023; 6:743. [PMID: 37463969 PMCID: PMC10354059 DOI: 10.1038/s42003-023-05118-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 07/07/2023] [Indexed: 07/20/2023] Open
Abstract
Blood phagocytes, such as neutrophils and monocytes, generate reactive oxygen species (ROS) as a part of host defense response against infections. We investigated the mechanism of Fcγ-Receptor (FcγR) mediated ROS production in these cells to understand how they contribute to anti-malarial immunity. Plasmodium falciparum merozoites opsonized with naturally occurring IgG triggered both intracellular and extracellular ROS generation in blood phagocytes, with neutrophils being the main contributors. Using specific inhibitors, we show that both FcγRIIIB and FcγRIIA acted synergistically to induce ROS production in neutrophils, and that NADPH oxidase 2 and the PI3K intracellular signal transduction pathway were involved in this process. High levels of neutrophil ROS were also associated with protection against febrile malaria in two geographically diverse malaria endemic regions from Ghana and India, stressing the importance of the cooperation between anti-malarial IgG and neutrophils in triggering ROS-mediated parasite killing as a mechanism for naturally acquired immunity against malaria.
Collapse
Affiliation(s)
- Ebenezer Addo Ofori
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Asier Garcia-Senosiain
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Mohammad Naghizadeh
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Ikhlaq Hussain Kana
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Morten Hanefeld Dziegiel
- Blood Bank KI 2034, Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Bright Adu
- Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Subhash Singh
- ICMR-Regional Medical Research Centre, Bhubaneswar, Odisha, India.
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
7
|
B-Cell Epitope Mapping of the Plasmodium falciparum Malaria Vaccine Candidate GMZ2.6c in a Naturally Exposed Population of the Brazilian Amazon. Vaccines (Basel) 2023; 11:vaccines11020446. [PMID: 36851323 PMCID: PMC9966924 DOI: 10.3390/vaccines11020446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
The GMZ2.6c malaria vaccine candidate is a multi-stage P. falciparum chimeric protein that contains a fragment of the sexual-stage Pfs48/45-6C protein genetically fused to GMZ2, an asexual-stage vaccine construction consisting of the N-terminal region of the glutamate-rich protein (GLURP) and the C-terminal region of the merozoite surface protein-3 (MSP-3). Previous studies showed that GMZ2.6c is widely recognized by antibodies from Brazilian exposed individuals and that its components are immunogenic in natural infection by P. falciparum. In addition, anti-GMZ2.6c antibodies increase with exposure to infection and may contribute to parasite immunity. Therefore, identifying epitopes of proteins recognized by antibodies may be an important tool for understanding protective immunity. Herein, we identify and validate the B-cell epitopes of GMZ2.6c as immunogenic and immunodominant in individuals exposed to malaria living in endemic areas of the Brazilian Amazon. Specific IgG antibodies and subclasses against MSP-3, GLURP, and Pfs48/45 epitopes were detected by ELISA using synthetic peptides corresponding to B-cell epitopes previously described for MSP-3 and GLURP or identified by BepiPred for Pfs48/45. The results showed that the immunodominant epitopes were P11 from GLURP and MSP-3c and DG210 from MSP-3. The IgG1 and IgG3 subclasses were preferentially induced against these epitopes, supporting previous studies that these proteins are targets for cytophilic antibodies, important for the acquisition of protective immunity. Most individuals presented detectable IgG antibodies against Pfs48/45a and/or Pfs48/45b, validating the prediction of linear B-cell epitopes. The higher frequency and antibody levels against different epitopes from GLURP, MSP-3, and Pfs48/45 provide additional information that may suggest the relevance of GMZ2.6c as a multi-stage malaria vaccine candidate.
Collapse
|
8
|
Kyei-Baafour E, Kusi KA, Arthur FK, Tiendrebeogo RW, Owusu-Yeboa E, Singh SK, Friedrich S, Gerds TA, Dodoo D, Theisen M, Adu B. High opsonic phagocytosis activity and growth inhibition of merozoites are associated with RON4 antibody levels and protect against febrile malaria in Ghanaian children. Front Immunol 2023; 14:1161301. [PMID: 37197657 PMCID: PMC10183564 DOI: 10.3389/fimmu.2023.1161301] [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: 02/08/2023] [Accepted: 04/17/2023] [Indexed: 05/19/2023] Open
Abstract
Background Naturally acquired immunity to malaria may involve different immune mechanisms working in concert, however, their respective contributions and potential antigenic targets have not been clearly established. Here, we assessed the roles of opsonic phagocytosis and antibody-mediated merozoite growth inhibition in Plasmodium falciparum (P. falciparum) infection outcomes in Ghanaian children. Methods The levels of merozoite opsonic phagocytosis, growth inhibition activities and six P. falciparum antigen-specific IgG of plasma samples from children (n=238, aged 0.5 to 13 years) were measured at baseline prior to the malaria seasons in southern Ghana. The children were then actively and passively followed up for febrile malaria and asymptomatic P. falciparum infection detection in a 50-week longitudinal cohort. P. falciparum infection outcome was modelled as a function of the measured immune parameters while accounting for important demographic factors. Results High plasma activity of opsonic phagocytosis [adjusted odds ratio (aOR)= 0.16; 95%CI= 0.05 - 0.50, p = 0.002], and growth inhibition (aOR=0.15; 95% CI = 0.04-0.47; p = 0.001) were individually associated with protection against febrile malaria. There was no evidence of correlation (b= 0.13; 95% CI= -0.04-0.30; p=0.14) between the two assays. IgG antibodies against MSPDBL1 correlated with opsonic phagocytosis (OP) while IgG against PfRh2a correlated with growth inhibition. Notably, IgG antibodies against RON4 correlated with both assays. Conclusion Opsonic phagocytosis and growth inhibition are protective immune mechanisms against malaria that may be acting independently to confer overall protection. Vaccines incorporating RON4 may benefit from both immune mechanisms.
Collapse
Affiliation(s)
- Eric Kyei-Baafour
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon Accra, Ghana
- Department of Biochemistry and Biotechnology, College of Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Kwadwo Asamoah Kusi
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon Accra, Ghana
| | - Fareed K.N. Arthur
- Department of Biochemistry and Biotechnology, College of Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Regis W. Tiendrebeogo
- Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Eunice Owusu-Yeboa
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon Accra, Ghana
| | - Susheel K. Singh
- Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Sarah Friedrich
- Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
- Department of Medical Statistics, University Medical Center Goettingen, Goettingen, Germany
| | - Thomas A. Gerds
- Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Daniel Dodoo
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon Accra, Ghana
| | - Michael Theisen
- Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Bright Adu
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon Accra, Ghana
- *Correspondence: Bright Adu,
| |
Collapse
|
9
|
Chan JA, Loughland JR, de la Parte L, Okano S, Ssewanyana I, Nalubega M, Nankya F, Musinguzi K, Rek J, Arinaitwe E, Tipping P, Bourke P, Andrew D, Dooley N, SheelaNair A, Wines BD, Hogarth PM, Beeson JG, Greenhouse B, Dorsey G, Kamya M, Hartel G, Minigo G, Feeney M, Jagannathan P, Boyle MJ. Age-dependent changes in circulating Tfh cells influence development of functional malaria antibodies in children. Nat Commun 2022; 13:4159. [PMID: 35851033 PMCID: PMC9293980 DOI: 10.1038/s41467-022-31880-6] [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: 12/12/2021] [Accepted: 07/08/2022] [Indexed: 01/29/2023] Open
Abstract
T-follicular helper (Tfh) cells are key drivers of antibodies that protect from malaria. However, little is known regarding the host and parasite factors that influence Tfh and functional antibody development. Here, we use samples from a large cross-sectional study of children residing in an area of high malaria transmission in Uganda to characterize Tfh cells and functional antibodies to multiple parasites stages. We identify a dramatic re-distribution of the Tfh cell compartment with age that is independent of malaria exposure, with Th2-Tfh cells predominating in early childhood, while Th1-Tfh cell gradually increase to adult levels over the first decade of life. Functional antibody acquisition is age-dependent and hierarchical acquired based on parasite stage, with merozoite responses followed by sporozoite and gametocyte antibodies. Antibodies are boosted in children with current infection, and are higher in females. The children with the very highest antibody levels have increased Tfh cell activation and proliferation, consistent with a key role of Tfh cells in antibody development. Together, these data reveal a complex relationship between the circulating Tfh compartment, antibody development and protection from malaria.
Collapse
Affiliation(s)
- Jo-Anne Chan
- Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Jessica R Loughland
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
- Global and Tropical Health Division, Menzies School of Health Research, Tiwi, Australia
| | | | - Satomi Okano
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Isaac Ssewanyana
- Infectious Diseases Research Collaboration, Kampala, Uganda
- London School of Hygiene and Tropical Medicine, London, UK
| | - Mayimuna Nalubega
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
- Infectious Diseases Research Collaboration, Kampala, Uganda
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | | | | | - John Rek
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Peta Tipping
- Global and Tropical Health Division, Menzies School of Health Research, Tiwi, Australia
| | - Peter Bourke
- Division of Medicine, Cairns Hospital, Manunda, QLD, Australia
| | - Dean Andrew
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Nicholas Dooley
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
- Griffith University, Brisbane, QLD, Australia
| | - Arya SheelaNair
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Bruce D Wines
- Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - P Mark Hogarth
- Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - James G Beeson
- Burnet Institute, Melbourne, VIC, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
- Department of Microbiology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | | | - Grant Dorsey
- University of California San Francisco, San Francisco, CA, USA
| | - Moses Kamya
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Gunter Hartel
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Gabriela Minigo
- Global and Tropical Health Division, Menzies School of Health Research, Tiwi, Australia
- College of Health and Human Sciences, Charles Darwin University, Darwin, NT, Australia
| | - Margaret Feeney
- University of California San Francisco, San Francisco, CA, USA
| | | | - Michelle J Boyle
- Burnet Institute, Melbourne, VIC, Australia.
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia.
- Global and Tropical Health Division, Menzies School of Health Research, Tiwi, Australia.
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.
- Griffith University, Brisbane, QLD, Australia.
| |
Collapse
|
10
|
Gonzales SJ, Clarke KN, Batugedara G, Garza R, Braddom AE, Reyes RA, Ssewanyana I, Garrison KC, Ippolito GC, Greenhouse B, Bol S, Bunnik EM. A Molecular Analysis of Memory B Cell and Antibody Responses Against Plasmodium falciparum Merozoite Surface Protein 1 in Children and Adults From Uganda. Front Immunol 2022; 13:809264. [PMID: 35720313 PMCID: PMC9201334 DOI: 10.3389/fimmu.2022.809264] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 05/05/2022] [Indexed: 01/18/2023] Open
Abstract
Memory B cells (MBCs) and plasma antibodies against Plasmodium falciparum (Pf) merozoite antigens are important components of the protective immune response against malaria. To gain understanding of how responses against Pf develop in these two arms of the humoral immune system, we evaluated MBC and antibody responses against the most abundant merozoite antigen, full-length Pf merozoite surface protein 1 (PfMSP1FL), in individuals from a region in Uganda with high Pf transmission. Our results showed that PfMSP1FL-specific B cells in adults with immunological protection against malaria were predominantly IgG+ classical MBCs, while children with incomplete protection mainly harbored IgM+ PfMSP1FL-specific classical MBCs. In contrast, anti-PfMSP1FL plasma IgM reactivity was minimal in both children and adults. Instead, both groups showed high plasma IgG reactivity against PfMSP1FL, with broadening of the response against non-3D7 strains in adults. The B cell receptors encoded by PfMSP1FL-specific IgG+ MBCs carried high levels of amino acid substitutions and recognized relatively conserved epitopes on the highly variable PfMSP1 protein. Proteomics analysis of PfMSP119-specific IgG in plasma of an adult revealed a limited repertoire of anti-MSP1 antibodies, most of which were IgG1 or IgG3. Similar to B cell receptors of PfMSP1FL-specific MBCs, anti-PfMSP119 IgGs had high levels of amino acid substitutions and their sequences were predominantly found in classical MBCs, not atypical MBCs. Collectively, these results showed evolution of the PfMSP1-specific humoral immune response with cumulative Pf exposure, with a shift from IgM+ to IgG+ B cell memory, diversification of B cells from germline, and stronger recognition of PfMSP1 variants by the plasma IgG repertoire.
Collapse
Affiliation(s)
- S. Jake Gonzales
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Kathleen N. Clarke
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Gayani Batugedara
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Rolando Garza
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Ashley E. Braddom
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Raphael A. Reyes
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Isaac Ssewanyana
- Infectious Disease Research Collaboration, Kampala, Uganda
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Kendra C. Garrison
- Department of Chemical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Gregory C. Ippolito
- Department of Molecular Biosciences and Department of Oncology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
| | - Bryan Greenhouse
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Sebastiaan Bol
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Evelien M. Bunnik
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
11
|
Knudsen AS, Walker MR, Agullet JP, Björnsson KH, Bassi MR, Barfod L. Enhancing neutralization of Plasmodium falciparum using a novel monoclonal antibody against the rhoptry-associated membrane antigen. Sci Rep 2022; 12:3040. [PMID: 35197516 PMCID: PMC8866459 DOI: 10.1038/s41598-022-06921-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 01/31/2022] [Indexed: 11/09/2022] Open
Abstract
The pathogenesis of malaria is associated with blood-stage infection and there is strong evidence that antibodies specific to parasite blood-stage antigens can control parasitemia. This provides a strong rational for applying blood-stage antigen components in a multivalent vaccine, as the induced antibodies in combination can enhance protection. The Plasmodium falciparum rhoptry-associated membrane antigen (PfRAMA) is a promising vaccine target, due to its fundamental role in merozoite invasion and low level of polymorphism. Polyclonal antibodies against PfRAMA are able to inhibit P. falciparum growth and interact synergistically when combined with antibodies against P. falciparum reticulocyte-binding protein 5 (PfRh5) or cysteine-rich protective antigen (PfCyRPA). In this study, we identified a novel PfRAMA-specific mAb with neutralizing activity, which in combination with PfRh5- or PfCyRPA-specific mAbs potentiated the neutralizing effect. By applying phage display technology, we mapped the protective epitope to be in the C-terminal region of PfRAMA. Our results confirmed previous finding of synergy between PfRAMA-, PfRh5- and PfCyRPA-specific antibodies, thereby paving the way of testing these antigens (or fragments of these antigens) in combination to improve the efficacy of blood-stage malaria vaccines. The results emphasize the importance of directing antibody responses towards protective epitopes, as the majority of anti-PfRAMA mAbs were unable to inhibit merozoite invasion of erythrocytes.
Collapse
Affiliation(s)
- Anne S Knudsen
- Department of Immunology and Microbiology, Centre for Medical Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Melanie R Walker
- Department of Immunology and Microbiology, Centre for Medical Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Judit P Agullet
- Department of Immunology and Microbiology, Centre for Medical Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kasper H Björnsson
- Department of Immunology and Microbiology, Centre for Medical Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maria R Bassi
- Department of Immunology and Microbiology, Centre for Medical Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lea Barfod
- Department of Immunology and Microbiology, Centre for Medical Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
12
|
Opi DH, Kurtovic L, Chan JA, Horton JL, Feng G, Beeson JG. Multi-functional antibody profiling for malaria vaccine development and evaluation. Expert Rev Vaccines 2021; 20:1257-1272. [PMID: 34530671 DOI: 10.1080/14760584.2021.1981864] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION A vaccine would greatly accelerate current global efforts toward malaria elimination. While a partially efficacious vaccine has been achieved for Plasmodium falciparum, a major bottleneck in developing highly efficacious vaccines is a lack of reliable correlates of protection, and the limited application of assays that quantify functional immune responses to evaluate and down-select vaccine candidates in pre-clinical studies and clinical trials. AREAS COVERED In this review, we describe the important role of antibodies in immunity against malaria and detail the nature and functional activities of antibodies against the malaria-causing parasite. We highlight the growing understanding of antibody effector functions against malaria and in vitro assays to measure these functional antibody responses. We discuss the application of these assays to quantify antibody functions in vaccine development and evaluation. EXPERT OPINION It is becoming increasingly clear that multiple antibody effector functions are involved in immunity to malaria. Therefore, we propose that evaluating vaccine candidates needs to move beyond individual assays or measuring IgG magnitude alone. Instead, vaccine evaluation should incorporate the full breadth of antibody response types and harness a wider range of assays measuring functional antibody responses. We propose a 3-tier approach to implementing assays to inform vaccine evaluation.
Collapse
Affiliation(s)
- D Herbert Opi
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - Liriye Kurtovic
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia
| | - Jo-Anne Chan
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - Jessica L Horton
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - Gaoqian Feng
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - James G Beeson
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia.,Department of Microbiology, Monash University, Clayton, Australia
| |
Collapse
|
13
|
O'Flaherty K, Oo WH, Zaloumis SG, Cutts JC, Aung KZ, Thein MM, Drew DR, Razook Z, Barry AE, Parischa N, Zaw NN, Thu HK, Thi A, Htay WYM, Soe AP, Simpson JA, Beeson JG, Agius PA, Fowkes FJI. Community-based molecular and serological surveillance of subclinical malaria in Myanmar. BMC Med 2021; 19:121. [PMID: 34044836 PMCID: PMC8161608 DOI: 10.1186/s12916-021-01993-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 04/27/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND In the Greater Mekong Subregion (GMS), current malaria surveillance strategies rely on a network of village health volunteers (VHVs) reporting the results of rapid diagnostic tests (RDTs), known to miss many asymptomatic infections. Integration of more sensitive diagnostic molecular and serological measures into the VHV network may improve surveillance of residual malaria transmission in hard-to-reach areas in the region and inform targeted interventions and elimination responses. However, data on residual malaria transmission that would be captured by these measures in the VHV-led testing and treatment surveillance network in the GMS is unknown. METHODS A total of 114 VHVs were trained to collect dried blood spots from villagers undergoing routine RDTs as part of VHV-led active and passive case detection from April 2015 to June 2016. Samples were subjected to molecular testing (quantitative polymerase chain reaction [qPCR]) to determine Plasmodium falciparum and P. vivax infection and serological testing (against P. falciparum and P. vivax antigens) to determine exposure to P. falciparum and P. vivax. RESULTS Over 15 months, 114 VHVs performed 32,194 RDTs and collected samples for molecular (n = 13,157) and serological (n = 14,128) testing. The prevalence of molecular-detectable P. falciparum and P. vivax infection was 3.2% compared to the 0.16% prevalence of Plasmodium spp. by RDT, highlighting the large burden of infections undetected by standard surveillance. Peaks in anti-P. falciparum, but not P. vivax, merozoite IgG seroprevalence coincided with seasonal P. falciparum transmission peaks, even in those with no molecularly detectable parasites. At the individual level, antibody seropositivity was associated with reduced odds of contemporaneous P. falciparum (OR for PfCSP 0.51 [95%CI 0.35, 0.76], p = 0.001, PfAMA1 0.70 [95%CI 0.52, 0.93], p = 0.01, and PfMSP2 0.81 [95%CI 0.61, 1.08], p = 0.15), but not P. vivax infection (OR PvAMA1 1.02 [95%CI 0.73, 1.43], p = 0.89) indicating a potential role of immunity in protection against molecular-detectable P. falciparum parasitaemia. CONCLUSIONS We demonstrated that integration and implementation of sample collection for molecular and serological surveillance into networks of VHV servicing hard-to-reach populations in the GMS is feasible, can capture significant levels of ongoing undetected seasonal malaria transmission and has the potential to supplement current routine RDT testing. Improving malaria surveillance by advancing the integration of molecular and serological techniques, through centralised testing approaches or novel point-of-contact tests, will advance progress, and tracking, towards malaria elimination goals in the GMS.
Collapse
Affiliation(s)
- Katherine O'Flaherty
- Burnet Institute for Medical Research and Public Health, Melbourne, Australia.,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - Win Han Oo
- Burnet Institute Myanmar, Yangon, Myanmar
| | - Sophie G Zaloumis
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - Julia C Cutts
- Burnet Institute for Medical Research and Public Health, Melbourne, Australia.,Department of Medicine, University of Melbourne, Melbourne, Australia
| | | | | | - Damien R Drew
- Burnet Institute for Medical Research and Public Health, Melbourne, Australia
| | - Zahra Razook
- School of Medicine, Deakin University, Geelong, Australia
| | - Alyssa E Barry
- Burnet Institute for Medical Research and Public Health, Melbourne, Australia.,Department of Medicine, University of Melbourne, Melbourne, Australia.,School of Medicine, Deakin University, Geelong, Australia
| | - Naanki Parischa
- Burnet Institute for Medical Research and Public Health, Melbourne, Australia
| | | | | | - Aung Thi
- Department of Public Health, Myanmar Ministry of Health, Nay Pyi Taw, Myanmar
| | | | | | - Julie A Simpson
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia
| | - James G Beeson
- Burnet Institute for Medical Research and Public Health, Melbourne, Australia.,Department of Medicine, University of Melbourne, Melbourne, Australia.,Department of Microbiology and Central Clinical School, Monash University, Melbourne, Australia
| | - Paul A Agius
- Burnet Institute for Medical Research and Public Health, Melbourne, Australia.,Department of Epidemiology and Preventative Medicine, Monash University, Melbourne, Australia.,Judith Lumley Centre, La Trobe University, Melbourne, Australia
| | - Freya J I Fowkes
- Burnet Institute for Medical Research and Public Health, Melbourne, Australia. .,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia. .,Department of Epidemiology and Preventative Medicine, Monash University, Melbourne, Australia. .,Department of Infectious Diseases, Monash University, Melbourne, Australia.
| |
Collapse
|
14
|
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
|
15
|
Suau R, Vidal M, Aguilar R, Ruiz-Olalla G, Vázquez-Santiago M, Jairoce C, Nhabomba AJ, Gyan B, Dosoo D, Asante KP, Owusu-Agyei S, Campo JJ, Izquierdo L, Cavanagh D, Coppel RL, Chauhan V, Angov E, Dutta S, Gaur D, Beeson JG, Moncunill G, Dobaño C. RTS,S/AS01 E malaria vaccine induces IgA responses against CSP and vaccine-unrelated antigens in African children in the phase 3 trial. Vaccine 2020; 39:687-698. [PMID: 33358704 DOI: 10.1016/j.vaccine.2020.12.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 11/05/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND The evaluation of immune responses to RTS,S/AS01 has traditionally focused on immunoglobulin (Ig) G antibodies that are only moderately associated with protection. The role of other antibody isotypes that could also contribute to vaccine efficacy remains unclear. Here we investigated whether RTS,S/AS01E elicits antigen-specific serum IgA antibodies to the vaccine and other malaria antigens, and we explored their association with protection. METHODS Ninety-five children (age 5-17 months old at first vaccination) from the RTS,S/AS01E phase 3 clinical trial who received 3 doses of RTS,S/AS01E or a comparator vaccine were selected for IgA quantification 1 month post primary immunization. Two sites with different malaria transmission intensities (MTI) and clinical malaria cases and controls, were included. Measurements of IgA against different constructs of the circumsporozoite protein (CSP) vaccine antigen and 16 vaccine-unrelated Plasmodium falciparum antigens were performed using a quantitative suspension array assay. RESULTS RTS,S vaccination induced a 1.2 to 2-fold increase in levels of serum/plasma IgA antibodies to all CSP constructs, which was not observed upon immunization with a comparator vaccine. The IgA response against 13 out of 16 vaccine-unrelated P. falciparum antigens also increased after vaccination, and levels were higher in recipients of RTS,S than in comparators. IgA levels to malaria antigens before vaccination were more elevated in the high MTI than the low MTI site. No statistically significant association of IgA with protection was found in exploratory analyses. CONCLUSIONS RTS,S/AS01E induces IgA responses in peripheral blood against CSP vaccine antigens and other P. falciparum vaccine-unrelated antigens, similar to what we previously showed for IgG responses. Collectively, data warrant further investigation of the potential contribution of vaccine-induced IgA responses to efficacy and any possible interplay, either synergistic or antagonistic, with protective IgG, as identifying mediators of protection by RTS,S/AS01E immunization is necessary for the design of improved second-generation vaccines. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov: NCT008666191.
Collapse
Affiliation(s)
- Roger Suau
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Rosselló 153 CEK Building, E-08036 Barcelona, Catalonia, Spain.
| | - Marta Vidal
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Rosselló 153 CEK Building, E-08036 Barcelona, Catalonia, Spain.
| | - Ruth Aguilar
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Rosselló 153 CEK Building, E-08036 Barcelona, Catalonia, Spain.
| | - Gemma Ruiz-Olalla
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Rosselló 153 CEK Building, E-08036 Barcelona, Catalonia, Spain.
| | - Miquel Vázquez-Santiago
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Rosselló 153 CEK Building, E-08036 Barcelona, Catalonia, Spain.
| | - Chenjerai Jairoce
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Rosselló 153 CEK Building, E-08036 Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929 Maputo, Mozambique.
| | - Augusto J Nhabomba
- Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929 Maputo, Mozambique
| | - Ben Gyan
- Noguchi Memorial Institute for Medical Research, University of Ghana, Ghana.
| | - David Dosoo
- Kintampo Health Research Centre, Kintampo, Ghana.
| | | | - Seth Owusu-Agyei
- Kintampo Health Research Centre, Kintampo, Ghana; Disease Control Department. London School of Hygiene and Tropical Medicine, London, UK
| | - Joseph J Campo
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Rosselló 153 CEK Building, E-08036 Barcelona, Catalonia, Spain.
| | - Luis Izquierdo
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Rosselló 153 CEK Building, E-08036 Barcelona, Catalonia, Spain.
| | - 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, Edinburgh, UK.
| | - Ross L Coppel
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, Victoria, Australia.
| | - Virander Chauhan
- 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 (WRAIR), Silver Spring, MD, USA.
| | - Sheetij Dutta
- U.S. Military Malaria Vaccine Program, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD, USA.
| | - 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
| | - James G Beeson
- Burnet Institute, Melbourne, Victoria, Australia; Central Clinical School, Monash University, Australia; Department of Medicine, University of Melbourne, Australia.
| | - Gemma Moncunill
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Rosselló 153 CEK Building, 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 CEK Building, E-08036 Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929 Maputo, Mozambique.
| |
Collapse
|
16
|
Gonzales SJ, Reyes RA, Braddom AE, Batugedara G, Bol S, Bunnik EM. Naturally Acquired Humoral Immunity Against Plasmodium falciparum Malaria. Front Immunol 2020; 11:594653. [PMID: 33193447 PMCID: PMC7658415 DOI: 10.3389/fimmu.2020.594653] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/07/2020] [Indexed: 12/18/2022] Open
Abstract
Malaria remains a significant contributor to the global burden of disease, with around 40% of the world's population at risk of Plasmodium infections. The development of an effective vaccine against the malaria parasite would mark a breakthrough in the fight to eradicate the disease. Over time, natural infection elicits a robust immune response against the blood stage of the parasite, providing protection against malaria. In recent years, we have gained valuable insight into the mechanisms by which IgG acts to prevent pathology and inhibit parasite replication, as well as the potential role of immunoglobulin M (IgM) in these processes. Here, we discuss recent advances in our understanding of the mechanisms, acquisition, and maintenance of naturally acquired immunity, and the relevance of these discoveries for the development of a potential vaccine against the blood stage of Plasmodium falciparum.
Collapse
Affiliation(s)
| | | | | | | | | | - Evelien M. Bunnik
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
17
|
Stoute JA, Landmesser ME, Biryukov S. Treatment of Plasmodium falciparum merozoites with the protease inhibitor E64 and mechanical filtration increases their susceptibility to complement activation. PLoS One 2020; 15:e0237786. [PMID: 32822376 PMCID: PMC7442247 DOI: 10.1371/journal.pone.0237786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 06/22/2020] [Indexed: 11/18/2022] Open
Abstract
Plasmodium falciparum malaria killed 451,000 people in 2017. Merozoites, the stage of the parasite that invades RBCs, are a logical target for vaccine development. Treatment with the protease inhibitor E64 followed by filtration through a 1.2 μm filter is being used to purify merozoites for immunologic assays. However, there have been no studies to determine the effect of these treatments on the susceptibility of merozoites to complement or antibodies. To address this gap, we purified merozoites with or without E64 followed by filtration through either a 1.2 or 2.7 μm filter, or no filtration. Merozoites were then incubated in either 10% fresh or heat-inactivated serum followed by surface staining and flow cytometry with monoclonal antibodies against the complement effector molecules C3b or C5b9. To determine the effect of anti-merozoite antibodies, we incubated merozoites with MAb5.2, a mouse monoclonal antibody that targets the merozoite surface protein 1. We used an amine-reactive fluorescent dye to measure membrane integrity. Treatment with E64 resulted in an insignificant increase in the proportion of merozoites that were C3b positive but in a significant increase in the proportion that were C5b9 positive. Filtration increased the proportion of merozoites that were either C3b or C5b9-positive. The combination of filtration and E64 treatment resulted in marked deposition of C3b and C5b9. MAb5.2 induced greater complement deposition than serum alone or an IgG2b isotype control. The combination of E64 treatment, filtration, and MAb5.2 resulted in very rapid and significant deposition of C5b9. Filtration through the 1.2 μm filter selected a population of merozoites with greater membrane integrity, but their integrity deteriorated rapidly upon exposure to serum. We conclude that E64 treatment and filtration increase the susceptibility of merozoites to complement and antibody. Filtered or E64-treated merozoites are not suitable for immunologic studies that address the efficacy of antibodies in vitro.
Collapse
Affiliation(s)
- José A. Stoute
- The Division of Infectious Diseases, Department of Medicine, the Penn State College of Medicine, Hershey, Pennsylvania, United States of America
- Department of Microbiology and Immunology, The Penn State College of Medicine, Hershey, Pennsylvania, United States of America
- * E-mail: ,
| | - Mary E. Landmesser
- The Division of Infectious Diseases, Department of Medicine, the Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Sergei Biryukov
- Department of Microbiology and Immunology, The Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| |
Collapse
|
18
|
Nagaoka H, Kanoi BN, Ntege EH, Aoki M, Fukushima A, Tsuboi T, Takashima E. Antibodies against a short region of PfRipr inhibit Plasmodium falciparum merozoite invasion and PfRipr interaction with Rh5 and SEMA7A. Sci Rep 2020; 10:6573. [PMID: 32313230 PMCID: PMC7171142 DOI: 10.1038/s41598-020-63611-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 03/30/2020] [Indexed: 12/29/2022] Open
Abstract
Plasmodium falciparum merozoite invasion into erythrocytes is an essential step of the blood-stage cycle, survival of parasites, and malaria pathogenesis. P. falciparum merozoite Rh5 interacting protein (PfRipr) forms a complex with Rh5 and CyRPA in sequential molecular events leading to erythrocyte invasion. Recently we described PfRipr as a conserved protein that induces strain-transcending growth inhibitory antibodies in in vitro assays. However, being a large and complex protein of 1086 amino acids (aa) with 87 cysteine residues, PfRipr is difficult to express in conventional expression systems towards vaccine development. In this study we sought to identify the most potent region of PfRipr that could be developed to overcome difficulties related to protein expression, as well as to elucidate the invasion inhibitory mechanism of anti-PfRipr antibodies. Using the wheat germ cell-free system, Ecto- PfRipr and truncates of approximately 200 aa were expressed as soluble proteins. We demonstrate that antibodies against PfRipr truncate 5 (PfRipr_5: C720-D934), a region within the PfRipr C-terminal EGF-like domains, potently inhibit merozoite invasion. Furthermore, the antibodies strongly block PfRipr/Rh5 interaction, as well as that between PfRipr and its erythrocyte-surface receptor, SEMA7A. Taken together, PfRipr_5 is a potential candidate for further development as a blood-stage malaria vaccine.
Collapse
Affiliation(s)
- Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, 3 Bunkyo-cho, Matsuyama, Japan
| | - Bernard N Kanoi
- Division of Malaria Research, Proteo-Science Center, Ehime University, 3 Bunkyo-cho, Matsuyama, Japan
| | - Edward H Ntege
- Division of Malaria Research, Proteo-Science Center, Ehime University, 3 Bunkyo-cho, Matsuyama, Japan.,Department of Plastic and Reconstructive Surgery, University of the Ryukyus, School of Medicine and Hospital, Okinawa, Japan
| | - Masamitsu Aoki
- Sumitomo Dainippon Pharma Co., Ltd, 3-1-98, Kasugadenaka, Konohanaku, Osaka, 554-0022, Japan
| | - Akihisa Fukushima
- Sumitomo Dainippon Pharma Co., Ltd, 3-1-98, Kasugadenaka, Konohanaku, Osaka, 554-0022, Japan
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, 3 Bunkyo-cho, Matsuyama, Japan
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, 3 Bunkyo-cho, Matsuyama, Japan.
| |
Collapse
|
19
|
Kana IH, Singh SK, Garcia-Senosiain A, Dodoo D, Singh S, Adu B, Theisen M. Breadth of Functional Antibodies Is Associated With Plasmodium falciparum Merozoite Phagocytosis and Protection Against Febrile Malaria. J Infect Dis 2020; 220:275-284. [PMID: 30820557 DOI: 10.1093/infdis/jiz088] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/26/2019] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The specific targets of functional antibodies against Plasmodium falciparum merozoites remain largely unexplored and, more importantly, their relevance to naturally acquired immunity in longitudinal cohort studies (LCSs) is yet to be tested. METHODS Functionality of immunoglobulin G (IgG) antibodies against 24 merozoite antigens was determined at the baseline of an LCS in Ghana using a bead-based opsonic phagocytosis assay (BPA). Antigen-specific IgG3 subclass antibodies were quantified in the same samples by the Luminex multiplex system. RESULTS A wide range of BPA activity was observed across the different antigens. High BPA responses of nMSP3K1, GLURP-R2, MSP23D7, MSP119k, and PfRh2-2030 coupled beads were significantly associated with a higher probability of children not experiencing febrile malaria. Children with high breadth of functional antibodies against these antigens together with cMSP33D7 had a significantly reduced risk of febrile malaria (adjusted hazard ratio, 0.36 [95% confidence interval, .18-.72]; P = .004). Five of the 6 BPA activities significantly (likelihood ratio rest, P ≤ .05) contributed to the protective immunity observed with the IgG3 antibodies. CONCLUSIONS The development of BPA allowed profiling of functional antibodies in an LCS. Identification of targets of opsonic phagocytosis may have implications in the development of a subunit malaria vaccine.
Collapse
Affiliation(s)
- Ikhlaq Hussain Kana
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Susheel Kumar Singh
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Asier Garcia-Senosiain
- Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Daniel Dodoo
- Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon
| | | | - Bright Adu
- Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon
| | - 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 and Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
20
|
Blank A, Fürle K, Jäschke A, Mikus G, Lehmann M, Hüsing J, Heiss K, Giese T, Carter D, Böhnlein E, Lanzer M, Haefeli WE, Bujard H. Immunization with full-length Plasmodium falciparum merozoite surface protein 1 is safe and elicits functional cytophilic antibodies in a randomized first-in-human trial. NPJ Vaccines 2020; 5:10. [PMID: 32025341 PMCID: PMC6994672 DOI: 10.1038/s41541-020-0160-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/14/2020] [Indexed: 12/20/2022] Open
Abstract
A vaccine remains a priority in the global fight against malaria. Here, we report on a single-center, randomized, double-blind, placebo and adjuvant-controlled, dose escalation phase 1a safety and immunogenicity clinical trial of full-length Plasmodium falciparum merozoite surface protein 1 (MSP1) in combination with GLA-SE adjuvant. Thirty-two healthy volunteers were vaccinated at least three times with MSP1 plus adjuvant, adjuvant alone, or placebo (24:4:4) to evaluate the safety and immunogenicity. MSP1 was safe, well tolerated and immunogenic, with all vaccinees sero-converting independent of the dose. The MSP1-specific IgG and IgM titers persisted above levels found in malaria semi-immune humans for at least 6 months after the last immunization. The antibodies were variant- and strain-transcending and stimulated respiratory activity in granulocytes. Furthermore, full-length MSP1 induced memory T-cells. Our findings encourage challenge studies as the next step to evaluate the efficacy of full-length MSP1 as a vaccine candidate against falciparum malaria (EudraCT 2016-002463-33).
Collapse
Affiliation(s)
- Antje Blank
- Klinische Pharmakologie und Pharmakoepidemiologie, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Kristin Fürle
- Center of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Anja Jäschke
- Center of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Gerd Mikus
- Klinische Pharmakologie und Pharmakoepidemiologie, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Monika Lehmann
- Koordinierungszentrum für Klinische Studien (KKS), Universitätsklinikum Heidelberg, Im Neuenheimer Feld 130.3, 69120 Heidelberg, Germany
| | - Johannes Hüsing
- Koordinierungszentrum für Klinische Studien (KKS), Universitätsklinikum Heidelberg, Im Neuenheimer Feld 130.3, 69120 Heidelberg, Germany
| | - Kirsten Heiss
- PEPperPRINT GmbH, Rischerstrasse 12, 69123 Heidelberg, Germany
| | - Thomas Giese
- Institut für Immunologie, Universitätsklinikum Heidelberg und Deutsches Zentrum für Infektionsforschung (DZIF) Standort Heidelberg, Im Neuenheimer Feld 305, 69120 Heidelberg, Germany
| | - Darrick Carter
- PAI Life Sciences, 1616 Eastlake Ave E, Suite 550, Seattle, WA 98102 USA
| | - Ernst Böhnlein
- Sumaya Biotech GmbH & Co. KG, Im Neuenheimer Feld 582, 69120 Heidelberg, Germany
| | - Michael Lanzer
- Center of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Walter E. Haefeli
- Klinische Pharmakologie und Pharmakoepidemiologie, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Hermann Bujard
- Sumaya Biotech GmbH & Co. KG, Im Neuenheimer Feld 582, 69120 Heidelberg, Germany
- Zentrum für Molekulare Biologie Heidelberg, Universität Heidelberg, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
| |
Collapse
|
21
|
Kochayoo P, Changrob S, Wangriatisak K, Lee SK, Chootong P, Han ET. The persistence of naturally acquired antibodies and memory B cells specific to rhoptry proteins of Plasmodium vivax in patients from areas of low malaria transmission. Malar J 2019; 18:382. [PMID: 31783870 PMCID: PMC6884809 DOI: 10.1186/s12936-019-3009-2] [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: 06/04/2019] [Accepted: 11/16/2019] [Indexed: 01/07/2023] Open
Abstract
Background Rhoptries are the large, paired, secretory organelles located at the apical tip of the malaria merozoite that are considered important for parasite invasion processes. Plasmodium vivax rhoptry proteins have been shown to induce humoral immunity during natural infections. Therefore, these proteins may be potential novel vaccine candidates. However, there is a lack of data on the duration of antibody and memory B cell (MBC) responses. Here, the longitudinal analysis of antibody and MBC responses to the P. vivax rhoptry proteins PvRALP1-Ecto and PvRhopH2 were monitored and analysed in individuals to determine their persistence. Methods Thirty-nine samples from P. vivax-infected subjects (age 18–60 years) were recruited to explore the frequency and persistence of antibody and MBC responses against rhoptry proteins (PvRALP1-Ecto and PvRhopH2) using both cross-sectional and longitudinal cohort study designs. Antibody levels were determined by ELISA during clinical malaria, and at 3, 9 and 12 months post-infection. The frequency of MBC sub-sets and presence of rhoptry-specific MBCs in subjects 18 months after treatment were detected by flow cytometry and ELISPOT assay. Results The seroprevalence of antibodies against PvRALP1-Ecto and PvRhopH2 proteins was found to be high during acute infection, with IgG1, IgG2 and IgG3 sub-classes predominant. However, these anti-rhoptry responses were short-lived and significantly decreased at 9 months post-infection. To relate the durability of these antibody responses to MBC persistence at post-infection, 18-month post-infection peripheral blood mononuclear cells (PBMCs) samples were taken to detect rhoptry-specific MBCs and frequency of MBC sub-sets, and correlate with antibody responses. These late post-infection samples revealed that rhoptry-specific MBCs were present in about 70% of total subjects. However, the persistence of specific MBCs was not correlated with antibody responses as the majority of malaria subjects who were positive for PvRALP1-Ecto- or PvRhopH2-specific MBCs were seronegative for the rhoptry antigens. The frequencies of classical MBCs were increased after infection, whereas those of activated and atypical MBCs were decreased, indicating that MBC responses could switch from activated or atypical MBCs to classical MBCs after parasite clearance, and were maintained in blood circulating at post-infection. Conclusion The study showed that rhoptry antigens induced the development and persistence of MBC responses in P. vivax-infected subjects who lived in a region of low malaria transmission, which were not related to the longevity of antibody responses.
Collapse
Affiliation(s)
- Piyawan Kochayoo
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Siriruk Changrob
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Kittikorn Wangriatisak
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Seong Kyun Lee
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 200-701, Republic of Korea
| | - Patchanee Chootong
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand.
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 200-701, Republic of Korea.
| |
Collapse
|
22
|
Farias E, Bezerra F, Baia-da-Silva DC, Chaves YO, Cardoza TB, de Almeida MEM, Oliveira LB, Lalwani P, Orlandi PP, Lacerda MVG, Lopes SCP, Nogueira PA. A simple, ex vivo phagocytosis assay of Plasmodium vivax merozoites by flow cytometry. Mem Inst Oswaldo Cruz 2019; 114:e190158. [PMID: 31596312 PMCID: PMC6783134 DOI: 10.1590/0074-02760190158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/04/2019] [Indexed: 11/22/2022] Open
Abstract
As phagocytosis is the first line of defense against malaria, we developed a
phagocytosis assay with Plasmodium vivax (P.
vivax) merozoites that can be applied to evaluate vaccine
candidates. Briefly, after leukocyte removal with loosely packed cellulose
powder in a syringe, P. vivax trophozoites matured to the
merozoite-rich schizont stages in the presence of the E64 protease inhibitor.
The Percoll gradient-enriched schizonts were chemically disrupted to release
merozoites that were submitted to merozoite opsonin-dependent phagocytosis in
two phagocytic lines with human and mouse antibodies against the N- and
C-terminus of P. vivax Merozoite Surface Protein-1
(Nterm-PvMSP1 and MSP119). The resulting assay is simple and
efficient for use as a routine phagocytic assay for the evaluation of merozoite
stage vaccine candidates.
Collapse
Affiliation(s)
- Elizangela Farias
- Universidade Federal do Amazonas, Programa de Pós-Graduação Stricto Sensu em Imunologia Básica e Aplicada, Manaus, AM, Brasil.,Fundação Oswaldo Cruz-Fiocruz, Instituto Leônidas e Maria Deane, Programa de Pós-Graduação Stricto Sensu em Biologia da Interação Patógeno Hospedeiro, Manaus, AM, Brasil
| | - Fhabiane Bezerra
- Universidade Federal do Amazonas, Programa de Pós-Graduação Stricto Sensu em Imunologia Básica e Aplicada, Manaus, AM, Brasil
| | | | - Yury Oliveira Chaves
- Fundação Oswaldo Cruz-Fiocruz, Instituto Leônidas e Maria Deane, Programa de Pós-Graduação Stricto Sensu em Biologia da Interação Patógeno Hospedeiro, Manaus, AM, Brasil.,Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Programa de Pós-Graduação Stricto Sensu em Biologia Parasitária, Rio de Janeiro, RJ, Brasil
| | - Tatiana Bacry Cardoza
- Fundação Oswaldo Cruz-Fiocruz, Instituto Leônidas e Maria Deane, Programa de Pós-Graduação Stricto Sensu em Biologia da Interação Patógeno Hospedeiro, Manaus, AM, Brasil
| | - Maria Edilene Martins de Almeida
- Fundação Oswaldo Cruz-Fiocruz, Instituto Leônidas e Maria Deane, Programa de Pós-Graduação Stricto Sensu em Biologia da Interação Patógeno Hospedeiro, Manaus, AM, Brasil.,Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Programa de Pós-Graduação Stricto Sensu em Biologia Celular e Molecular, Rio de Janeiro, RJ, Brasil
| | - Lucas Barbosa Oliveira
- Fundação Oswaldo Cruz-Fiocruz, Instituto Leônidas e Maria Deane, Programa de Pós-Graduação Stricto Sensu em Biologia da Interação Patógeno Hospedeiro, Manaus, AM, Brasil
| | - Pritesh Lalwani
- Universidade Federal do Amazonas, Programa de Pós-Graduação Stricto Sensu em Imunologia Básica e Aplicada, Manaus, AM, Brasil.,Fundação Oswaldo Cruz-Fiocruz, Instituto Leônidas e Maria Deane, Programa de Pós-Graduação Stricto Sensu em Biologia da Interação Patógeno Hospedeiro, Manaus, AM, Brasil
| | - Patrícia Puccinelli Orlandi
- Fundação Oswaldo Cruz-Fiocruz, Instituto Leônidas e Maria Deane, Programa de Pós-Graduação Stricto Sensu em Biologia da Interação Patógeno Hospedeiro, Manaus, AM, Brasil
| | - Marcus Vinicius Guimaraes Lacerda
- Fundação Oswaldo Cruz-Fiocruz, Instituto Leônidas e Maria Deane, Programa de Pós-Graduação Stricto Sensu em Biologia da Interação Patógeno Hospedeiro, Manaus, AM, Brasil.,Fundação de Medicina Tropical Dr Heitor Vieira Dourado, Manaus, AM, Brasil
| | - Stefanie Costa Pinto Lopes
- Fundação Oswaldo Cruz-Fiocruz, Instituto Leônidas e Maria Deane, Programa de Pós-Graduação Stricto Sensu em Biologia da Interação Patógeno Hospedeiro, Manaus, AM, Brasil
| | - Paulo Afonso Nogueira
- Fundação Oswaldo Cruz-Fiocruz, Instituto Leônidas e Maria Deane, Programa de Pós-Graduação Stricto Sensu em Biologia da Interação Patógeno Hospedeiro, Manaus, AM, Brasil
| |
Collapse
|
23
|
Kana IH, Garcia-Senosiain A, Singh SK, Tiendrebeogo RW, Chourasia BK, Malhotra P, Sharma SK, Das MK, Singh S, Adu B, Theisen M. Cytophilic Antibodies Against Key Plasmodium falciparum Blood Stage Antigens Contribute to Protection Against Clinical Malaria in a High Transmission Region of Eastern India. J Infect Dis 2019; 218:956-965. [PMID: 29733355 DOI: 10.1093/infdis/jiy258] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 05/02/2018] [Indexed: 12/21/2022] Open
Abstract
Background The collection of clinical data from a tribal population in a malaria-endemic area of India suggests the occurrence of naturally acquired immunity (NAI) against Plasmodium falciparum malaria. Methods Quantity and functionality of immunoglobulin G (IgG) antibodies against intact merozoites and recombinant proteins were assessed in a 13-month longitudinal cohort study of 121 individuals, 3-60 years of age. Results Opsonic phagocytosis of merozoites activity was strongly associated (hazard ratio [HR] = 0.34; 95% confidence interval [CI] = .18-.66; P = .0013) with protection against febrile malaria. Of the different IgG subclasses, only IgG3 antibodies against intact whole merozoites was significantly associated with protection against febrile malaria (HR = 0.47; 95% CI = .26-.86; P = .01). Furthermore, a combination of IgG3 antibody responses against Pf12, MSP3.7, MSP3.3, and MSP2FC27 was strongly associated with protection against febrile malaria (HR = 0.15; 95% CI, .06-.37; P = .0001). Conclusions These data suggest that NAI may, at least in part, be explained by opsonic phagocytosis of merozoites and IgG3 responses against whole merozoites, and in particular to a combination of 4 antigens is critical in this population. These results may have implications in the development of a subunit malaria vaccine. Opsonic phagocytosis of Plasmodium falciparum merozoites was associated with protection against clinical malaria in an India population. Antibody profiling identified four merozoite antigens (Pf12, MSP3.7, MSP3.3, and MSP2) as targets of protective Immunoglobuline G3 antibodies.
Collapse
Affiliation(s)
- Ikhlaq Hussain Kana
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Asier Garcia-Senosiain
- Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Susheel K Singh
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Régis Wendpayangde Tiendrebeogo
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Bishwanath Kumar Chourasia
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Pawan Malhotra
- Malaria Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Surya K Sharma
- National Institute of Malaria Research, Indian Council of Medical Research, New Delhi, India
| | - Manoj K Das
- National Institute of Malaria Research, Field Unit, Ranchi (Jharkhand), India
| | - Subhash Singh
- Indian Institute of Integrative Medicine, Canal Road, Jammu, India
| | - Bright Adu
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon
| | - 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 and Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Denmark
| |
Collapse
|
24
|
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
|
25
|
Mbengue B, Fall MM, Varela ML, Loucoubar C, Joos C, Fall B, Niang MS, Niang B, Mbow M, Dieye A, Perraut R. Analysis of antibody responses to selected Plasmodium falciparum merozoite surface antigens in mild and cerebral malaria and associations with clinical outcomes. Clin Exp Immunol 2019; 196:86-96. [PMID: 30580455 DOI: 10.1111/cei.13254] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2018] [Indexed: 11/28/2022] Open
Abstract
Merozoite surface proteins (MSPs) are critical for parasite invasion; they represent attractive targets for antibody-based protection against clinical malaria. To identify protection-associated target MSPs, the present study analysed antibody responses to whole merozoite extract (ME) and to defined MSP recombinant antigens in hospitalized patients from a low endemic urban area as a function of disease severity (mild versus cerebral malaria). Sera from 110 patients with confirmed severe cerebral malaria (CM) and 91 patients with mild malaria (MM) were analysed (mean age = 29 years) for total and subclass immunoglobulin (Ig)G to ME and total IgG to MSP1p19, MSP2, MSP3, MSP4 and MSP5 by enzyme-linked immunosorbent assay (ELISA). Functional antibody responses were evaluated using the antibody-dependent respiratory burst (ADRB) assay in a subset of sera. There was a trend towards higher IgG1 and IgG4 levels to ME in CM compared to MM; only ME IgM responses differed significantly between fatal and surviving CM patients. Increased prevalence of IgG to individual MSPs was found in the CM compared to the MM group, including significantly higher levels of IgG to MSP4 and MSP5 in the former. Sera from fatal (24·5%) versus surviving cases showed significantly lower IgG to MSP1p19 and MSP3 (P < 0·05). ADRB assay readouts correlated with high levels of anti-MSP IgG, and trended higher in sera from patients with surviving compared to fatal CM outcome (P = 0·07). These results document strong differential antibody responses to MSP antigens as targets of protective immunity against CM and in particular MSP1p19 and MSP3 as prognostic indicators.
Collapse
Affiliation(s)
- B Mbengue
- Service d'Immunologie FMPO, Université Cheikh Anta Diop de Dakar, Senegal.,Unité d'Immunogénétique, Institut Pasteur de Dakar, IPD, Senegal
| | - M M Fall
- Service de Réanimation, Hôpital Principal de Dakar, HPD, Senegal
| | - M-L Varela
- Unité d'Immunologie, Institut Pasteur de Dakar, IPD, Senegal
| | - C Loucoubar
- Groupe de Biostatistique et Bioinformatique, IPD, Senegal
| | - C Joos
- Unité d'Immunologie, Institut Pasteur de Dakar, IPD, Senegal
| | - B Fall
- Fédération des Laboratoires, Hôpital Principal de Dakar, HPD, Senegal
| | - M S Niang
- Service d'Immunologie FMPO, Université Cheikh Anta Diop de Dakar, Senegal
| | - B Niang
- Service de Réanimation, Hôpital Principal de Dakar, HPD, Senegal
| | - M Mbow
- Service d'Immunologie FMPO, Université Cheikh Anta Diop de Dakar, Senegal
| | - A Dieye
- Service d'Immunologie FMPO, Université Cheikh Anta Diop de Dakar, Senegal.,Unité d'Immunogénétique, Institut Pasteur de Dakar, IPD, Senegal
| | - R Perraut
- Unité d'Immunogénétique, Institut Pasteur de Dakar, IPD, Senegal.,Unité d'Immunologie, Institut Pasteur de Dakar, IPD, Senegal
| |
Collapse
|
26
|
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
|
27
|
Astawa INM, Oka IBM, Dwinata IM. Antibody immunoglobulin G1 and immunoglobulin G2a responses against some cystic fluid proteins of Cysticercus bovis in Balb/c mice. Vet World 2018; 11:1641-1647. [PMID: 30587902 PMCID: PMC6303487 DOI: 10.14202/vetworld.2018.1641-1647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 10/11/2018] [Indexed: 01/23/2023] Open
Abstract
Background and Aim: Immunoglobulin (Ig) G1 and IgG2a are the surrogate markers respectively for humoral and cellular immune responses of hosts against antigens including cystic fluid proteins of Cysticercus bovis. A study was conducted to investigate the IgG1 and IgG2a responses of Balb/c mice against some individual cystic fluid proteins of C. bovis in an effort to determine the roles of each protein in inducing the humoral and cellular immune responses in host. Materials and Methods: Individual p71, p31, and p14 proteins of C. bovis were purified by separation of the proteins using sodium dodecyl sulfate-polyacrylamide gel electrophoresis and elution of individual proteins from the gel. Six female Balb/c mice were immunized 4 times at 10-day intervals with the crude cystic fluid proteins, and sera were collected for the measurement of IgG1 and IgG2a levels against the individual proteins. Sera samples collected before the first immunization were used as negative antibody control, sera samples collected after the fourth immunization were used as positive antibody control, and crude cystic fluid protein was used as positive antigen control. Results: All immunized mice were immune to p71, p31, p14, and crude cystic fluid proteins of C. bovis. The crude cystic fluid proteins of C. bovis induced a higher IgG2a than IgG1 level following the first and the second immunizations but switched into a higher IgG1 than IgG2a level following the fourth immunization. Protein 71 kDa (p71) induced a higher IgG2a than IgG1 level following the fourth immunization. In contrast, p14 induced a higher IgG1 than IgG2a level following the fourth immunization. Low and balance IgG1 and IgG2a levels against p31 were observed following the first to the fourth immunizations. Conclusion: Using IgG1 and IgG2a levels as the surrogate markers, it appears that cystic fluid antigens of C. bovis induce both humoral and cellular immune responses in Balb/c mice. The p71 appears to be a better inducer of cellular immune response, whereas p14 is a better inducer of humoral immune response of mice.
Collapse
Affiliation(s)
- I Nyoman Mantik Astawa
- Laboratory of Immunology, Faculty of Veterinary Medicine, Udayana University, Denpasar Bali 80232, Indonesia
| | - Ida Bagus Made Oka
- Laboratory of Parasitology, Faculty of Veterinary Medicine, Udayana University, Bali 80232, Indonesia
| | - I Made Dwinata
- Laboratory of Parasitology, Faculty of Veterinary Medicine, Udayana University, Bali 80232, Indonesia
| |
Collapse
|
28
|
Frimpong A, Kusi KA, Ofori MF, Ndifon W. Novel Strategies for Malaria Vaccine Design. Front Immunol 2018; 9:2769. [PMID: 30555463 PMCID: PMC6281765 DOI: 10.3389/fimmu.2018.02769] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/12/2018] [Indexed: 12/19/2022] Open
Abstract
The quest for a licensed effective vaccine against malaria remains a global priority. Even though classical vaccine design strategies have been successful for some viral and bacterial pathogens, little success has been achieved for Plasmodium falciparum, which causes the deadliest form of malaria due to its diversity and ability to evade host immune responses. Nevertheless, recent advances in vaccinology through high throughput discovery of immune correlates of protection, lymphocyte repertoire sequencing and structural design of immunogens, provide a comprehensive approach to identifying and designing a highly efficacious vaccine for malaria. In this review, we discuss novel vaccine approaches that can be employed in malaria vaccine design.
Collapse
Affiliation(s)
- Augustina Frimpong
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.,Immunology Department, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana.,African Institute for Mathematical Sciences, Cape Coast, Ghana
| | - Kwadwo Asamoah Kusi
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.,Immunology Department, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Michael Fokuo Ofori
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.,Immunology Department, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Wilfred Ndifon
- African Institute for Mathematical Sciences, Cape Coast, Ghana.,African Institute for Mathematical Sciences, University of Stellenbosch, Cape Town, South Africa
| |
Collapse
|
29
|
Muh F, Lee SK, Hoque MR, Han JH, Park JH, Firdaus ER, Moon RW, Lau YL, Han ET. In vitro invasion inhibition assay using antibodies against Plasmodium knowlesi Duffy binding protein alpha and apical membrane antigen protein 1 in human erythrocyte-adapted P. knowlesi A1-H.1 strain. Malar J 2018; 17:272. [PMID: 30049277 PMCID: PMC6062950 DOI: 10.1186/s12936-018-2420-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 07/18/2018] [Indexed: 12/22/2022] Open
Abstract
Background The rapid process of malaria erythrocyte invasion involves ligand–receptor interactions. Inducing antibodies against specific ligands or receptors that abrogate the
invasion process is a key challenge for blood stage vaccine development. However, few candidates were reported and remain to be validated for the discovery of new vaccine candidates in Plasmodium knowlesi. Methods In order to investigate the efficacy of pre-clinical vaccine candidates in P. knowlesi-infected human cases, this study describes an in vitro invasion inhibition assay, using a P. knowlesi strain adapted to in vitro growth in human erythrocytes, PkA1-H.1. Recombinant proteins of P. knowlesi Duffy binding protein alpha (PkDBPα) and apical membrane antigen 1 (PkAMA1) were produced in Escherichia coli system and rabbit antibodies were generated from immune animals. Results PkDBPα and PkAMA1 recombinant proteins were expressed as insoluble and produced as a functional refolded form for this study. Antibodies against PkDBPα and PkAMA1 specifically recognized recombinant proteins and native parasite proteins in schizont-stage parasites on the merozoite organelles. Single and combination of anti-PkDBPα and anti-PkAMA1 antibodies elicited strong growth inhibitory effects on the parasite in concentration-dependent manner. Meanwhile, IgG prevalence of PkDBPα and PkAMA1 were observed in 13.0 and 46.7% in human clinical patients, respectively. Conclusion These data provide support for the validation of in vitro growth inhibition assay using antibodies of DBPα and AMA1 in human-adapted P. knowlesi parasite PkA1-H.1 strain. Electronic supplementary material The online version of this article (10.1186/s12936-018-2420-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fauzi Muh
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Seong-Kyun Lee
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Mohammad Rafiul Hoque
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Ji-Hoon Park
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Egy Rahman Firdaus
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Robert W Moon
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Yee Ling Lau
- Department of Parasitology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea.
| |
Collapse
|
30
|
Plasmodium falciparum MSP3 Exists in a Complex on the Merozoite Surface and Generates Antibody Response during Natural Infection. Infect Immun 2018; 86:IAI.00067-18. [PMID: 29760216 DOI: 10.1128/iai.00067-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/01/2018] [Indexed: 12/22/2022] Open
Abstract
Plasmodium falciparum merozoite surface protein 3 (MSP3) is an abundantly expressed secreted merozoite surface protein and a leading malaria vaccine candidate antigen. However, it is unclear how MSP3 is retained on the surface of merozoites without a glycosylphosphatidylinositol (GPI) anchor or a transmembrane domain. In the present study, we identified an MSP3-associated network on the Plasmodium merozoite surface by immunoprecipitation of Plasmodium merozoite lysate using antibody to the N terminus of MSP3 (anti-MSP3N) followed by mass spectrometry analysis. The results suggested the association of MSP3 with other merozoite surface proteins: MSP1, MSP6, MSP7, RAP2, and SERA5. Protein-protein interaction studies by enzyme-linked immunosorbent assay (ELISA) and surface plasmon resonance (SPR) analysis showed that MSP3 complex consists of MSP1, MSP6, and MSP7 proteins. Immunological characterization of MSP3 revealed that MSP3N is strongly recognized by hyperimmune serum from African and Asian populations. Furthermore, we demonstrate that human antibodies, affinity purified against recombinant MSP3N (rMSP3N), promote opsonic phagocytosis of merozoites in cooperation with monocytes. At nonphysiological concentrations, anti-MSP3N antibodies inhibited the growth of P. falciparum in vitro Together, the data suggest that MSP3 and especially its N-terminal region containing known B/T cell epitopes are targets of naturally acquired immunity against malaria and also comprise an important candidate for a multisubunit malaria vaccine.
Collapse
|
31
|
Lyth O, Vizcay-Barrena G, Wright KE, Haase S, Mohring F, Najer A, Henshall IG, Ashdown GW, Bannister LH, Drew DR, Beeson JG, Fleck RA, Moon RW, Wilson DW, Baum J. Cellular dissection of malaria parasite invasion of human erythrocytes using viable Plasmodium knowlesi merozoites. Sci Rep 2018; 8:10165. [PMID: 29976932 PMCID: PMC6033891 DOI: 10.1038/s41598-018-28457-z] [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: 05/16/2018] [Accepted: 06/22/2018] [Indexed: 12/21/2022] Open
Abstract
Plasmodium knowlesi, a zoonotic parasite causing severe-to-lethal malaria disease in humans, has only recently been adapted to continuous culture with human red blood cells (RBCs). In comparison with the most virulent human malaria, Plasmodium falciparum, there are, however, few cellular tools available to study its biology, in particular direct investigation of RBC invasion by blood-stage P. knowlesi merozoites. This leaves our current understanding of biological differences across pathogenic Plasmodium spp. incomplete. Here, we report a robust method for isolating viable and invasive P. knowlesi merozoites to high purity and yield. Using this approach, we present detailed comparative dissection of merozoite invasion (using a variety of microscopy platforms) and direct assessment of kinetic differences between knowlesi and falciparum merozoites. We go on to assess the inhibitory potential of molecules targeting discrete steps of invasion in either species via a quantitative invasion inhibition assay, identifying a class of polysulfonate polymer able to efficiently inhibit invasion in both, providing a foundation for pan-Plasmodium merozoite inhibitor development. Given the close evolutionary relationship between P. knowlesi and P. vivax, the second leading cause of malaria-related morbidity, this study paves the way for inter-specific dissection of invasion by all three major pathogenic malaria species.
Collapse
Affiliation(s)
- Oliver Lyth
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, UK
| | - Gema Vizcay-Barrena
- Centre for Ultrastructural Imaging, Guy's Campus, King's College London, London, UK
| | - Katherine E Wright
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, UK
| | - Silvia Haase
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, UK
| | - Franziska Mohring
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Adrian Najer
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, UK
| | - Isabelle G Henshall
- Research Centre for Infectious Diseases, School of Biological Sciences, The University of Adelaide, Adelaide, Australia
| | - George W Ashdown
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, UK
| | - Lawrence H Bannister
- Centre for Ultrastructural Imaging, Guy's Campus, King's College London, London, UK
| | - Damien R Drew
- Burnet Institute, 85 Commercial Road, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Victoria, Australia
| | - James G Beeson
- Burnet Institute, 85 Commercial Road, Melbourne, Victoria, Australia.,Central Clinical School, Monash University, Victoria, Australia
| | - Roland A Fleck
- Centre for Ultrastructural Imaging, Guy's Campus, King's College London, London, UK
| | - Robert W Moon
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Danny W Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, The University of Adelaide, Adelaide, Australia. .,Burnet Institute, 85 Commercial Road, Melbourne, Victoria, Australia.
| | - Jake Baum
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London, UK.
| |
Collapse
|
32
|
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
|
33
|
Merozoite Surface Protein 1 from Plasmodium falciparum Is a Major Target of Opsonizing Antibodies in Individuals with Acquired Immunity against Malaria. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00155-17. [PMID: 28877929 DOI: 10.1128/cvi.00155-17] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/31/2017] [Indexed: 11/20/2022]
Abstract
Naturally acquired immunity against malaria is largely mediated by serum antibodies controlling levels of blood-stage parasites. A limited understanding of the antigenic targets and functional mechanisms of protective antibodies has hampered the development of efficient malaria vaccines. Besides directly inhibiting the growth of Plasmodium parasites, antibodies can opsonize merozoites and recruit immune effector cells such as monocytes and neutrophils. Antibodies against the vaccine candidate merozoite surface protein 1 (MSP-1) are acquired during natural infections and have been associated with protection against malaria in several epidemiological studies. Here we analyzed serum antibodies from semi-immune individuals from Burkina Faso for their potential (i) to directly inhibit the growth of P. falciparum blood stages in vitro and (ii) to opsonize merozoites and to induce the antibody-dependent respiratory burst (ADRB) activity of neutrophils. While a few sera that directly inhibited the growth of P. falciparum blood stages were identified, immunoglobulin G (IgG) from all individuals clearly mediated the activation of neutrophils. The level of neutrophil activation correlated with levels of antibodies to MSP-1, and affinity-purified MSP-1-specific antibodies elicited ADRB activity. Furthermore, immunization of nonhuman primates with recombinant full-size MSP-1 induced antibodies that efficiently opsonized P. falciparum merozoites. Reversing the function by preincubation with recombinant antigens allowed us to quantify the contribution of MSP-1 to the antiparasitic effect of serum antibodies. Our data suggest that MSP-1, especially the partially conserved subunit MSP-183, is a major target of opsonizing antibodies acquired during natural exposure to malaria. Induction of opsonizing antibodies might be a crucial effector mechanism for MSP-1-based malaria vaccines.
Collapse
|
34
|
Perraut R, Varela ML, Joos C, Diouf B, Sokhna C, Mbengue B, Tall A, Loucoubar C, Touré A, Mercereau-Puijalon O. Association of antibodies to Plasmodium falciparum merozoite surface protein-4 with protection against clinical malaria. Vaccine 2017; 35:6720-6726. [PMID: 29042203 DOI: 10.1016/j.vaccine.2017.10.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 09/10/2017] [Accepted: 10/05/2017] [Indexed: 11/17/2022]
Abstract
Identification of parasite antigens targeted by immune effector mechanisms that confer protection against malaria is important for the design of a multi-component malaria vaccine. Here, the association of antibodies reacting with the Plasmodium falciparum merozoite surface protein-4 (MSP4) with protection against clinical malaria was investigated in a Senegalese community living in an area of moderate, seasonal malaria transmission. Blood samples were collected at the end of an 8-month long dry season without any recorded parasite transmission from 206 residents enrolled in a prospective follow-up study. Active daily clinical monitoring was implemented during the subsequent five months. Entomologic monitoring documented parasite transmission during the first three months of follow-up. Serum IgG levels were determined by ELISA against three MSP4 baculovirus-encoded recombinant protein constructs, namely the full-length MSP4p40, MSP4p30 devoid of a highly polymorphic sequence stretch and the conserved C-terminal EGF-containing MSP4p20, as well as against a merozoite crude extract. Community seroprevalence against all three constructs was quite high, the lowest being against MSP4p30. Seroprevalence and antibody levels against the three MSP4 constructs were age-dependent. IgG1 dominated the anti-MSP4p20 responses, while both IgG1 and IgG3 were observed against MSP4p40. Anti-MSP4 antibodies were associated with the antibody-dependent respiratory burst (ADRB) activity in a functional assay of merozoite phagocytosis by polymorphonuclear cells. Importantly, high antibody levels against each of the three MSP4 constructs at the end of the dry season were associated with reduced morbidity during the subsequent transmission season in an age-adjusted Poisson regression model (IRR = 0.65 [0.50-0.83], P<0.001 for responses over the median values). These data are consistent with a protective role for the naturally acquired anti-MSP4 antibodies and support further development of MSP4 as a candidate component of malaria vaccine.
Collapse
Affiliation(s)
- Ronald Perraut
- Unité d'Immunologie, Institut Pasteur de Dakar, Senegal.
| | | | | | - Babacar Diouf
- Unité d'Immunologie, Institut Pasteur de Dakar, Senegal
| | - Cheikh Sokhna
- Institut de Recherche pour le Développement (IRD), URMITE, UMR 198, Dakar, Senegal
| | | | - Adama Tall
- Unité d'Epidémiologie, Institut Pasteur de Dakar, Senegal
| | - Cheikh Loucoubar
- Institut Pasteur de Dakar, G4 Biostatistiques Bioinformatique et Modélisation, Dakar, Senegal
| | | | - Odile Mercereau-Puijalon
- Institut Pasteur, Département Parasites et Insectes Vecteurs, 25 Rue du Dr. Roux, 75015 Paris, France
| |
Collapse
|
35
|
França CT, White MT, He WQ, Hostetler JB, Brewster J, Frato G, Malhotra I, Gruszczyk J, Huon C, Lin E, Kiniboro B, Yadava A, Siba P, Galinski MR, Healer J, Chitnis C, Cowman AF, Takashima E, Tsuboi T, Tham WH, Fairhurst RM, Rayner JC, King CL, Mueller I. Identification of highly-protective combinations of Plasmodium vivax recombinant proteins for vaccine development. eLife 2017; 6:28673. [PMID: 28949293 PMCID: PMC5655538 DOI: 10.7554/elife.28673] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 09/25/2017] [Indexed: 12/22/2022] Open
Abstract
The study of antigenic targets of naturally-acquired immunity is essential to identify and prioritize antigens for further functional characterization. We measured total IgG antibodies to 38 P. vivax antigens, investigating their relationship with prospective risk of malaria in a cohort of 1–3 years old Papua New Guinean children. Using simulated annealing algorithms, the potential protective efficacy of antibodies to multiple antigen-combinations, and the antibody thresholds associated with protection were investigated for the first time. High antibody levels to multiple known and newly identified proteins were strongly associated with protection (IRR 0.44–0.74, p<0.001–0.041). Among five-antigen combinations with the strongest protective effect (>90%), EBP, DBPII, RBP1a, CyRPA, and PVX_081550 were most frequently identified; several of them requiring very low antibody levels to show a protective association. These data identify individual antigens that should be prioritized for further functional testing and establish a clear path to testing a multicomponent P. vivax vaccine.
Collapse
Affiliation(s)
- Camila Tenorio França
- Division of Population Health and Immunity, Walter and Eliza Hall Institute, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Michael T White
- Division of Population Health and Immunity, Walter and Eliza Hall Institute, Parkville, Australia.,MRC Center for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, Imperial College London, London, United Kingdom
| | - Wen-Qiang He
- Department of Medical Biology, University of Melbourne, Parkville, Australia.,Division of Infection and Immunity, Walter and Eliza Hall Institute, Parkville, Australia
| | - Jessica B Hostetler
- Malaria Programme, Wellcome Trust Sanger Institute, Hinxton, United Kingdom.,Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, United States
| | - Jessica Brewster
- Division of Infection and Immunity, Walter and Eliza Hall Institute, Parkville, Australia
| | - Gabriel Frato
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, United States
| | - Indu Malhotra
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, United States
| | - Jakub Gruszczyk
- Division of Infection and Immunity, Walter and Eliza Hall Institute, Parkville, Australia
| | - Christele Huon
- Malaria Parasite Biology and Vaccines Unit, Institut Pasteur, Paris, France
| | - Enmoore Lin
- Malaria Immuno-Epidemiology Unit, PNG Institute of Medical Research, Yagaum, Papua New Guinea
| | - Benson Kiniboro
- Malaria Immuno-Epidemiology Unit, PNG Institute of Medical Research, Yagaum, Papua New Guinea
| | - Anjali Yadava
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, United States
| | - Peter Siba
- Malaria Immuno-Epidemiology Unit, PNG Institute of Medical Research, Yagaum, Papua New Guinea
| | - Mary R Galinski
- International Center for Malaria Research, Education, and Development, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, United States.,Infectious Diseases Division, Department of Medicine, Emory University, Atlanta, United States
| | - Julie Healer
- Department of Medical Biology, University of Melbourne, Parkville, Australia.,Division of Infection and Immunity, Walter and Eliza Hall Institute, Parkville, Australia
| | - Chetan Chitnis
- Malaria Parasite Biology and Vaccines Unit, Institut Pasteur, Paris, France.,International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Alan F Cowman
- Department of Medical Biology, University of Melbourne, Parkville, Australia.,Division of Infection and Immunity, Walter and Eliza Hall Institute, Parkville, Australia
| | - Eizo Takashima
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, United States
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Wai-Hong Tham
- Department of Medical Biology, University of Melbourne, Parkville, Australia.,Division of Infection and Immunity, Walter and Eliza Hall Institute, Parkville, Australia
| | - Rick M Fairhurst
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, United States
| | - Julian C Rayner
- Malaria Programme, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Christopher L King
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, United States
| | - Ivo Mueller
- Division of Population Health and Immunity, Walter and Eliza Hall Institute, Parkville, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Australia.,Malaria Parasites and Hosts Unit, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France.,Barcelona Institute of Global Health, Barcelona, Spain
| |
Collapse
|
36
|
Hill DL, Schofield L, Wilson DW. IgG opsonization of merozoites: multiple immune mechanisms for malaria vaccine development. Int J Parasitol 2017; 47:585-595. [PMID: 28668325 DOI: 10.1016/j.ijpara.2017.05.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 04/12/2017] [Accepted: 05/12/2017] [Indexed: 02/07/2023]
Abstract
Global eradication of the human-infecting malaria parasite Plasmodium falciparum, the major cause of malaria mortality, is unlikely to be achieved without an effective vaccine. However, our limited understanding of how protective immune responses target malaria parasites in humans, and how to best elicit these immune responses through vaccination, has hampered vaccine development. The red blood cell invading stage of the parasite lifecycle (merozoite) displays antigens that are attractive vaccine candidates as they are accessible to antibodies and raise high antibody titres in naturally immune individuals. The number of merozoite antigens that elicit an immune response, and their structural and functional diversity, has led to a large number of lead antigens being pursued as vaccine candidates. Despite being seemingly spoilt for choice in terms of vaccine candidates, there is still a lack of consensus on exactly how merozoite antibodies reduce parasitemia and malaria disease. In this review we describe the various immune mechanisms that can result from IgG opsonization of merozoites, and highlight recent developments that support a role for these functional antibodies in naturally acquired and vaccine-induced immunity.
Collapse
Affiliation(s)
- Danika L Hill
- Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom; The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia.
| | - Louis Schofield
- Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, Australia
| | - Danny W Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, Australia; Burnet Institute, 85 Commercial Road, Melbourne 3004, Victoria, Australia.
| |
Collapse
|
37
|
Ntege EH, Takashima E, Morita M, Nagaoka H, Ishino T, Tsuboi T. Blood-stage malaria vaccines: post-genome strategies for the identification of novel vaccine candidates. Expert Rev Vaccines 2017; 16:769-779. [PMID: 28604122 DOI: 10.1080/14760584.2017.1341317] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION An efficacious malaria vaccine is necessary to advance the current control measures towards malaria elimination. To-date, only RTS,S/AS01, a leading pre-erythrocytic stage vaccine completed phase 3 trials, but with an efficacy of 28-36% in children, and 18-26% in infants, that waned over time. Blood-stage malaria vaccines protect against disease, and are considered effective targets for the logical design of next generation vaccines to improve the RTS,S field efficacy. Therefore, novel blood-stage vaccine candidate discovery efforts are critical, albeit with several challenges including, high polymorphisms in vaccine antigens, poor understanding of targets of naturally protective immunity, and difficulties in the expression of high AT-rich plasmodial proteins. Areas covered: PubMed ( www.ncbi.nlm.nih.gov/pubmed ) was searched to review the progress and future prospects of malaria vaccine research and development. We focused on post-genome vaccine candidate discovery, malaria vaccine development, sequence diversity, pre-clinical and clinical trials. Expert commentary: Post-genome high-throughput technologies using wheat germ cell-free protein synthesis technology and immuno-profiling with sera from malaria patients with clearly defined outcomes are highlighted to overcome current challenges of malaria vaccine candidate discovery.
Collapse
Affiliation(s)
- Edward H Ntege
- a Division of Malaria Research , Proteo-Science Center, Ehime University , Matsuyama , Ehime , Japan
| | - Eizo Takashima
- a Division of Malaria Research , Proteo-Science Center, Ehime University , Matsuyama , Ehime , Japan
| | - Masayuki Morita
- a Division of Malaria Research , Proteo-Science Center, Ehime University , Matsuyama , Ehime , Japan
| | - Hikaru Nagaoka
- a Division of Malaria Research , Proteo-Science Center, Ehime University , Matsuyama , Ehime , Japan
| | - Tomoko Ishino
- b Division of Molecular Parasitology , Proteo-Science Center, Ehime University , Toon , Ehime , Japan
| | - Takafumi Tsuboi
- a Division of Malaria Research , Proteo-Science Center, Ehime University , Matsuyama , Ehime , Japan
| |
Collapse
|
38
|
Loughland JR, Minigo G, Sarovich DS, Field M, Tipping PE, Montes de Oca M, Piera KA, Amante FH, Barber BE, Grigg MJ, William T, Good MF, Doolan DL, Engwerda CR, Anstey NM, McCarthy JS, Woodberry T. Plasmacytoid dendritic cells appear inactive during sub-microscopic Plasmodium falciparum blood-stage infection, yet retain their ability to respond to TLR stimulation. Sci Rep 2017; 7:2596. [PMID: 28572564 PMCID: PMC5453946 DOI: 10.1038/s41598-017-02096-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/05/2017] [Indexed: 12/13/2022] Open
Abstract
Plasmacytoid dendritic cells (pDC) are activators of innate and adaptive immune responses that express HLA-DR, toll-like receptor (TLR) 7, TLR9 and produce type I interferons. The role of human pDC in malaria remains poorly characterised. pDC activation and cytokine production were assessed in 59 malaria-naive volunteers during experimental infection with 150 or 1,800 P. falciparum-parasitized red blood cells. Using RNA sequencing, longitudinal changes in pDC gene expression were examined in five adults before and at peak-infection. pDC responsiveness to TLR7 and TLR9 stimulation was assessed in-vitro. Circulating pDC remained transcriptionally stable with gene expression altered for 8 genes (FDR < 0.07). There was no upregulation of co-stimulatory molecules CD86, CD80, CD40, and reduced surface expression of HLA-DR and CD123 (IL-3R-α). pDC loss from the circulation was associated with active caspase-3, suggesting pDC apoptosis during primary infection. pDC remained responsive to TLR stimulation, producing IFN-α and upregulating HLA-DR, CD86, CD123 at peak-infection. In clinical malaria, pDC retained HLA-DR but reduced CD123 expression compared to convalescence. These data demonstrate pDC retain function during a first blood-stage P. falciparum exposure despite sub-microscopic parasitaemia downregulating HLA-DR. The lack of evident pDC activation in both early infection and malaria suggests little response of circulating pDC to infection.
Collapse
Affiliation(s)
- Jessica R Loughland
- Menzies School of Health Research, Darwin, Australia and Charles Darwin University, Darwin, Australia.
| | - Gabriela Minigo
- Menzies School of Health Research, Darwin, Australia and Charles Darwin University, Darwin, Australia
| | - Derek S Sarovich
- Menzies School of Health Research, Darwin, Australia and Charles Darwin University, Darwin, Australia.,Centre for Animal Health Innovation, Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| | - Matt Field
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Peta E Tipping
- Menzies School of Health Research, Darwin, Australia and Charles Darwin University, Darwin, Australia.,Royal Darwin Hospital, Darwin, Australia
| | | | - Kim A Piera
- Menzies School of Health Research, Darwin, Australia and Charles Darwin University, Darwin, Australia
| | - Fiona H Amante
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Bridget E Barber
- Menzies School of Health Research, Darwin, Australia and Charles Darwin University, Darwin, Australia.,Infectious Diseases Unit, Queen Elizabeth Hospital, Kota Kinabalu, Sabah, Malaysia
| | - Matthew J Grigg
- Menzies School of Health Research, Darwin, Australia and Charles Darwin University, Darwin, Australia.,Infectious Diseases Unit, Queen Elizabeth Hospital, Kota Kinabalu, Sabah, Malaysia
| | - Timothy William
- Infectious Diseases Unit, Queen Elizabeth Hospital, Kota Kinabalu, Sabah, Malaysia.,Sabah Department of Health, Kota Kinabalu, Sabah, Malaysia
| | | | - Denise L Doolan
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia.,QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - Nicholas M Anstey
- Menzies School of Health Research, Darwin, Australia and Charles Darwin University, Darwin, Australia.,Royal Darwin Hospital, Darwin, Australia
| | | | - Tonia Woodberry
- Menzies School of Health Research, Darwin, Australia and Charles Darwin University, Darwin, Australia
| |
Collapse
|
39
|
Kana IH, Adu B, Tiendrebeogo RW, Singh SK, Dodoo D, Theisen M. Naturally Acquired Antibodies Target the Glutamate-Rich Protein on Intact Merozoites and Predict Protection Against Febrile Malaria. J Infect Dis 2017; 215:623-630. [PMID: 28329101 DOI: 10.1093/infdis/jiw617] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/05/2017] [Indexed: 11/14/2022] Open
Abstract
Background Plasmodium species antigens accessible at the time of merozoite release are likely targets of biologically functional antibodies. Methods Immunoglobulin G (IgG) antibodies against intact merozoites were quantified in the plasma of Ghanaian children from a longitudinal cohort using a novel flow cytometry-based immunofluorescence assay. Functionality of these antibodies, as well as glutamate-rich protein (GLURP)-specific affinity-purified IgG from malaria hyperimmune Liberian adults, was assessed by the opsonic phagocytosis (OP) assay. Results Opsonic phagocytosis activity was strongly associated (hazard ratio [HR] = 0.46; 95% confidence interval [CI] = .30-.73; P = .0008) with protection against febrile malaria. Of the antimerozoite-specific antibodies, only IgG3 was significantly associated with both OP and protection (HR = 0.53; 95% CI = .34-.84; Pcorrected = .03) against febrile malaria. Similarly, GLURP-specific antibodies previously shown to be protective against febrile malaria in this same cohort were significantly associated with OP activity in this study. GLURP-specific antibodies recognized merozoites and also mediated OP activity. Conclusions These findings support previous studies that found OP of merozoites to be associated with protection against malaria and further shows IgG3 and GLURP antibodies are key in the OP mechanism, thus giving further impetus for the development of malaria vaccines targeting GLURP.
Collapse
Affiliation(s)
- Ikhlaq Hussain Kana
- Department for Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark,Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen.,Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Bright Adu
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon
| | - Régis Wendpayangde Tiendrebeogo
- Department for Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark,Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen.,Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Susheel Kumar Singh
- Department for Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark,Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen.,Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Daniel Dodoo
- Noguchi Memorial Institute for Medical Research, University of Ghana, Legon
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark,Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen.,Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|