1
|
Dieppois C, Adjemout M, Cretin J, Gallardo F, Torres M, Picard C, Sawadogo SA, Rihet P, Paul P. An Expression Quantitative Trait Locus of Fc Gamma Receptor Genes Is Associated With Antimalarial IgG Responses and Infection Levels in Burkinabe Families. J Infect Dis 2025; 231:1008-1019. [PMID: 39450553 DOI: 10.1093/infdis/jiae528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/01/2024] [Accepted: 10/22/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND The interaction between antibodies and Fcγ receptors (FcγRs) plays a critical role in regulating immune responses to Plasmodium falciparum. Polymorphisms in genes encoding FcγRs influence the host's capacity to control parasite infection. This study investigates whether noncoding variants influencing FcγR expression are associated with antimalarial immunization and infection traits. METHODS We utilized eQTL databases and functional annotations to identify noncoding variants, specifically rs1771575, rs2099684, and rs6700241, within the FCGR gene cluster. In addition, we examined the coding variants rs1801274 (p.His167Arg) and rs1050501 (p.Ile231Thr), which affect the affinity of FcγRIIa and FcγRIIb for IgG. These variants were genotyped in 163 individuals from Burkinabe families. Family-based linear mixed regression and Quantitative Transmission Disequilibrium Tests (QTDT) analyses were performed to assess associations with IgG levels and malaria infection, accounting for relevant covariates. RESULTS Linear mixed models identified rs1771575 as associated with total IgG levels, while both rs1771575 and rs1801274 were linked to IgG2, and rs1050501 to IgG1 levels. A haplotype combining rs2099684 and rs6700241 was positively associated with IgG1. The rs1771575-CC and rs1050501-TT genotypes correlated with higher infection levels in children. QTDT models confirmed the association of rs1771575 with IgG2 and infection in children. CONCLUSIONS Our findings suggest that the intergenic variant rs1771575 serves as an independent marker for IgG levels and blood infection in children. This highlights the interplay between regulatory variants and coding mutations in FCGR, which may influence immune function and antibody production. These results underscore the potential for personalized strategies to monitor humoral responses in malaria-endemic regions.
Collapse
Affiliation(s)
- Christelle Dieppois
- Theories and Approaches of Genomic Complexity, Aix Marseille University, INSERM U1090, Marseille, France
| | - Mathieu Adjemout
- Theories and Approaches of Genomic Complexity, Aix Marseille University, INSERM U1090, Marseille, France
| | - Jules Cretin
- Theories and Approaches of Genomic Complexity, Aix Marseille University, INSERM U1090, Marseille, France
| | - Frederic Gallardo
- Theories and Approaches of Genomic Complexity, Aix Marseille University, INSERM U1090, Marseille, France
| | - Magali Torres
- Theories and Approaches of Genomic Complexity, Aix Marseille University, INSERM U1090, Marseille, France
| | - Christophe Picard
- Anthropologie bio-culturelle, droit, éthique et santé, Établissement Français du Sang, Centre National de la Recherche Scientifique, Aix Marseille University, Marseille, France
- Immunogenetics Laboratory, Etablissement Français du Sang, Marseille, France
| | - Serge Aimé Sawadogo
- Unité de Formation en Sciences de la Santé, Université Joseph Ki-Zerbo, Ouagadougou, Burkina Faso
- Centre Pour la Pecherche et l'Pnnovation en Pmmunologie Pédicale de Puagadougou-Nelson Mandela, Ouagadougou, Burkina Faso
| | - Pascal Rihet
- Theories and Approaches of Genomic Complexity, Aix Marseille University, INSERM U1090, Marseille, France
| | - Pascale Paul
- Theories and Approaches of Genomic Complexity, Aix Marseille University, INSERM U1090, Marseille, France
| |
Collapse
|
2
|
Hammi I, Giron-Michel J, Akarid K, Arnoult D. FcRγIIA response duality in leishmaniasis. Microb Pathog 2025; 198:107123. [PMID: 39557223 DOI: 10.1016/j.micpath.2024.107123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/25/2024] [Accepted: 11/14/2024] [Indexed: 11/20/2024]
Abstract
Leishmania is responsible for a neglected tropical disease affecting millions of people around the world and could potentially spread more due to climate change. This disease not only leads to significant morbidity but also imposes substantial social and economic burdens on affected populations, often exacerbating poverty and health disparities. Despite the complexity and effectiveness of the immune response, the parasite has developed various strategies to evade detection and manipulates host cells in favor of its replication. These evasion strategies start at early stages of the infection by hijacking immune receptors to silence critical cellular response that would otherwise limit the pathogen's propagation. Among these receptors, Fc receptors have emerged as a significant player in the immune evasion strategies employed by microorganisms, as they could promote inhibitory pathways. This review explores the potential role of one of these immune receptors, the FcγRIIA, in leishmaniasis and how this parasite may use it and the signaling pathways downstream to evade the host immune response. By understanding the potential interactions between Leishmania and immune receptors such as FcγRIIA, we may identify novel targets for therapeutic intervention aimed to enhance the host immune response and reduce the burden of this disease.
Collapse
Affiliation(s)
- Ikram Hammi
- Health & Environment Laboratory, Ain Chock Faculty of Sciences, Hassan II University of Casablanca (UH2C), Morocco; INSERM UMR-S-MD 1197, Ministère des Armées et Université Paris Saclay, Villejuif, France.
| | - Julien Giron-Michel
- INSERM UMR-S-MD 1197, Ministère des Armées et Université Paris Saclay, Villejuif, France
| | - Khadija Akarid
- Health & Environment Laboratory, Ain Chock Faculty of Sciences, Hassan II University of Casablanca (UH2C), Morocco
| | - Damien Arnoult
- INSERM UMR-S-MD 1197, Ministère des Armées et Université Paris Saclay, Villejuif, France.
| |
Collapse
|
3
|
Hammi I, Giron-Michel J, Riyad M, Akarid K, Arnoult D. FcRγIIA attenuates pathology of cutaneous leishmaniasis and modulates ITAMa/i balance. Parasit Vectors 2024; 17:517. [PMID: 39696675 DOI: 10.1186/s13071-024-06593-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/19/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Leishmania is the causal parasite of leishmaniasis, a neglected tropical disease affecting millions of individuals worldwide, and its dissemination is linked to climate change. Despite the complexity and effectiveness of the immune response, the parasite has developed many strategies to evade it and take control of the host cell to replicate. These evasion strategies start at early stages of infection by hijacking immune receptors to mitigate the cellular response. In this study, we examined whether Leishmania uses the Fc receptor FcγRIIA/CD32a and its downstream signaling pathways to evade the host immune response. METHODS Regarding in vivo studies, CD32a transgenic mice and the corresponding wild types were infected with Leishmania major Friedlin strain. For the in vitro experiments, BMDMs isolated from WT or CD32a transgenic mice and control or CD32a knockdown differentiated THP-1s were infected with two species of Leishmania, Leishmania major and L. tropica. RESULTS In vivo, expression of FcγRIIA/CD32a was found to accelerate the signs of inflammation while simultaneously preventing the formation of necrotic lesions after Leishmania infection. In infected macrophages, the presence of FcγRIIA/CD32a did not affect the secretion of proinflammatory cytokines, while the balance between ITAMa and ITAMi proteins was disturbed with improved Fyn and Lyn activation. Unexpectedly, infection with L. tropica but not L. major triggered an intracytoplasmic processing of FcγRIIA/CD32a. CONCLUSIONS Our observations underscore the significance of FcγRIIA/CD32a in cutaneous leishmaniasis and its potential use as a therapeutic target.
Collapse
Affiliation(s)
- Ikram Hammi
- Health & Environment Laboratory, Ain Chock Faculty of Sciences, Hassan II University of Casablanca (UH2C), Casablanca, Morocco
- INSERM UMR-S-MD 1197, Ministère des Armées et Université Paris Saclay, Villejuif, France
| | - Julien Giron-Michel
- INSERM UMR-S-MD 1197, Ministère des Armées et Université Paris Saclay, Villejuif, France
| | - Myriam Riyad
- Laboratory of Cellular and Molecular Pathology, Faculty of Medicine and Pharmacy, UH2C, Casablanca, Morocco
| | - Khadija Akarid
- Health & Environment Laboratory, Ain Chock Faculty of Sciences, Hassan II University of Casablanca (UH2C), Casablanca, Morocco
| | - Damien Arnoult
- INSERM UMR-S-MD 1197, Ministère des Armées et Université Paris Saclay, Villejuif, France.
| |
Collapse
|
4
|
Cretin J, Adjemout M, Dieppois C, Gallardo F, Torres M, Merard Z, Sawadogo SA, Picard C, Rihet P, Paul P. A Non-Coding Fc Gamma Receptor Cis-Regulatory Variant within the 1q23 Gene Cluster Is Associated with Plasmodium falciparum Infection in Children Residing in Burkina Faso. Int J Mol Sci 2023; 24:15711. [PMID: 37958695 PMCID: PMC10650193 DOI: 10.3390/ijms242115711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/27/2023] [Accepted: 08/31/2023] [Indexed: 11/15/2023] Open
Abstract
Antibodies play a crucial role in activating protective immunity against malaria by interacting with Fc-gamma receptors (FcγRs). Genetic variations in genes encoding FcγRs can affect immune cell responses to the parasite. In this study, our aim was to investigate whether non-coding variants that regulate FcγR expression could influence the prevalence of Plasmodium falciparum infection. Through bioinformatics approaches, we selected expression quantitative trait loci (eQTL) for FCGR2A, FCGR2B, FCGR2C, FCGR3A, and FCGR3B genes encoding FcγRs (FCGR), in whole blood. We prioritized two regulatory variants, rs2099684 and rs1771575, located in open genomic regions. These variants were identified using RegVar, ImmuNexUT, and transcription factor annotations specific to immune cells. In addition to these, we genotyped the coding variants FCGR2A/rs1801274 and FCGR2B/rs1050501 in 234 individuals from a malaria-endemic area in Burkina Faso. We conducted age and family-based analyses to evaluate associations with the prevalence of malarial infection in both children and adults. The analysis revealed that the regulatory rs1771575-CC genotype was predicted to influence FCGR2B/FCGR2C/FCGR3A transcripts in immune cells and was the sole variant associated with a higher prevalence of malarial infection in children. In conclusion, this study identifies the rs1771575 cis-regulatory variant affecting several FcγRs in myeloid and neutrophil cells and associates it with the inter-individual capacity of children living in Burkina Faso to control malarial infection.
Collapse
Affiliation(s)
- Jules Cretin
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
- Institut MarMaRa, 13288 Marseille, France
| | - Mathieu Adjemout
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
- Institut MarMaRa, 13288 Marseille, France
| | - Christelle Dieppois
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
| | - Frederic Gallardo
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
| | - Magali Torres
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
| | - Zachary Merard
- ADES UMR, Aix Marseille University, 13288 Marseille, France (C.P.)
| | - Serge Aimé Sawadogo
- Unité de Formation en Sciences de la Santé (UFR/SDS), Université Joseph KI-ZERBO, Ouagadougou 03 BP 7021, Burkina Faso;
- Centre PrïmO-Nelson Mandela, 84 rue Sao Tomé et Principe, Ouagadougou 09 BP 706, Burkina Faso
| | - Christophe Picard
- ADES UMR, Aix Marseille University, 13288 Marseille, France (C.P.)
- Immunogenetics Laboratory, Etablissement Français du Sang PACA-Corse, 13001 Marseille, France
| | - Pascal Rihet
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
| | - Pascale Paul
- INSERM 1090, TAGC Theories and Approaches of Genomic Complexity, Campus de Luminy, Aix Marseille University, 13288 Marseille, France (M.A.); (C.D.); (F.G.); (M.T.)
| |
Collapse
|
5
|
Tayipto Y, Liu Z, Mueller I, Longley RJ. Serology for Plasmodium vivax surveillance: A novel approach to accelerate towards elimination. Parasitol Int 2021; 87:102492. [PMID: 34728377 DOI: 10.1016/j.parint.2021.102492] [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] [Received: 03/31/2021] [Revised: 08/30/2021] [Accepted: 10/28/2021] [Indexed: 01/13/2023]
Abstract
Plasmodium vivax is the most widespread causative agent of human malaria in the world. Despite the ongoing implementation of malaria control programs, the rate of case reduction has declined over the last 5 years. Hence, surveillance of malaria transmission should be in place to identify and monitor areas that require intensified malaria control interventions. Serological tools may offer additional insights into transmission intensity over parasite and entomological measures, especially as transmission levels decline. Antibodies can be detected in the host system for months to even years after parasite infections have been cleared from the blood, enabling malaria exposure history to be captured. Because the Plasmodium parasite expresses more than 5000 proteins, it is important to a) understand antibody longevity following infection and b) measure antibodies to more than one antigen in order to accurately inform on the exposure and/or immune status of populations. This review summarises current practices for surveillance of P. vivax malaria, the current state of research into serological exposure markers and their potential role for accelerating malaria elimination, and discusses further studies that need to be undertaken to see such technology implemented in malaria-endemic areas.
Collapse
Affiliation(s)
- Yanie Tayipto
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Zoe Liu
- The Institute for Mental and Physical Health and Clinical Translation, Barwon Health, Deakin University, Geelong, Victoria, Australia; School of Medicine, Centre for Molecular and Medical Research, Deakin University, Geelong, Australia
| | - Ivo Mueller
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Rhea J Longley
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
6
|
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
|
7
|
Factor VIII Fc Fusion Protein but not FVIII Drives Human Monocyte-Derived Dendritic Cell Activation via FcγRIIa. Hemasphere 2020; 4:e330. [PMID: 32072146 PMCID: PMC7000470 DOI: 10.1097/hs9.0000000000000330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022] Open
Abstract
This study compares the effect of recombinant Factor VIII Fc fusion protein (rFVIII-Fc) with recombinant FVIII (rFVIII) on monocyte-derived dendritic cells (moDC's). Cells treated with rFVIII-Fc showed morphological changes typical for cell activation, had a significant up-regulation of cell activation markers and produced higher levels of pro-inflammatory cytokines. Even after stimulation with Lipopolysaccharides, the addition of rFVIII-Fc led to increased expression of activation markers, indicating that rFVIII-Fc is capable of amplifying the maturation signal. On the contrary, cultivation of moDC's with rFVIII did not alter cell morphology or increase surface activation marker expression and pro-inflammatory cytokine production. The binding of the Fc domain to the activating Fcγ receptor IIa (FcγRIIa) can cause cell activation. Therefore, the effect of rFVIII-Fc on FcγRIIa was analyzed in detail. Cultivation of moDC's with rFVIII-Fc led to increased phosphorylation of FcγRIIa, which was not detected for rFVIII. Blocking FcγRIIa prior to the cultivation with rFVIII-Fc significantly reduced the activating effect of rFVIII-Fc, indicating that rFVIII-Fc-induced moDC activation was caused by FcγRIIa. Moreover, rFVIII-Fc bound to FCGR2A-transfected human embryonic kidney 293 cells. Taken together, our data present a new mechanism of moDC activation by rFVIII-Fc via FcγRIIa.
Collapse
|
8
|
Kumagai T, Palacios A, Casadevall A, García MJ, Toro C, Tiemeyer M, Prados-Rosales R. Serum IgM Glycosylation Associated with Tuberculosis Infection in Mice. mSphere 2019; 4:e00684-18. [PMID: 30918063 PMCID: PMC6437276 DOI: 10.1128/msphere.00684-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/07/2019] [Indexed: 01/14/2023] Open
Abstract
Changes in serum glycans discriminate between disease statuses in cancer. A similar connection has not been established in the context of infectious diseases such as tuberculosis (TB). The inflammation arising from infection by Mycobacterium tuberculosis may affect host protein glycosylation, thereby providing information about disease status in TB. A mouse model of infection was used to study glycoprotein N-glycosylation in serum. Following digestion of serum glycoproteins with peptide-N-glycosidase F (PNGase F), released glycans were permethylated and analyzed by multidimensional mass spectrometry (MS). Conditions included naive or Mycobacterium bovis BCG-vaccinated animals, which were either uninfected or infected with M. tuberculosis MS results were validated by lectin blotting. We found that both glycoprotein fucosylation and sialylation were particularly sensitive to M. tuberculosis infection. We observed that M. tuberculosis infection elevates serum IgM levels and induces changes in glycosylation that could inform about the disease.IMPORTANCE We demonstrate that M. tuberculosis infection influenced host protein glycosylation in a mouse model. The mechanism by which infection modifies glycans in serum proteins is not understood. Investigation of the regulation of such modifications by M. tuberculosis opens a new field that could lead to the discovery of novel biomarkers. Validation of such findings in human samples will reveal the clinical relevance of these findings.
Collapse
Affiliation(s)
- Tadahiro Kumagai
- Complex Carbohydrate Research Center, The University of Georgia, Athens, Georgia, USA
| | | | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - M Jesús García
- Department of Preventive Medicine, Public Health and Microbiology, Autonomous University of Madrid, Madrid, Spain
| | - Carlos Toro
- Service of Microbiology, Hospital Universitario La Paz, IdiPaz, Madrid, Spain
| | - Michael Tiemeyer
- Complex Carbohydrate Research Center, The University of Georgia, Athens, Georgia, USA
| | - Rafael Prados-Rosales
- CIC bioGUNE, Derio, Bizkaia, Spain
- Department of Preventive Medicine, Public Health and Microbiology, Autonomous University of Madrid, Madrid, Spain
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
9
|
Lee JH, Ko K. Production of Recombinant Anti-Cancer Vaccines in Plants. Biomol Ther (Seoul) 2017; 25:345-353. [PMID: 28554196 PMCID: PMC5499611 DOI: 10.4062/biomolther.2016.126] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 12/13/2016] [Accepted: 02/06/2017] [Indexed: 12/24/2022] Open
Abstract
Plant expression systems have been developed to produce anti-cancer vaccines. Plants have several advantages as bioreactors for the production of subunit vaccines: they are considered safe, and may be used to produce recombinant proteins at low production cost. However, several technical issues hinder large-scale production of anti-cancer vaccines in plants. The present review covers design strategies to enhance the immunogenicity and therapeutic potency of anti-cancer vaccines, methods to increase vaccine-expressing plant biomass, and challenges facing the production of anti-cancer vaccines in plants. Specifically, the issues such as low expression levels and plant-specific glycosylation are described, along with their potential solutions.
Collapse
Affiliation(s)
- Jeong Hwan Lee
- Department of Medicine, Therapeutic Protein Engineering Lab, College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Kisung Ko
- Department of Medicine, Therapeutic Protein Engineering Lab, College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
10
|
Virus-Like Particle (VLP) Plus Microcrystalline Tyrosine (MCT) Adjuvants Enhance Vaccine Efficacy Improving T and B Cell Immunogenicity and Protection against Plasmodium berghei/vivax. Vaccines (Basel) 2017; 5:vaccines5020010. [PMID: 28468322 PMCID: PMC5492007 DOI: 10.3390/vaccines5020010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/18/2017] [Accepted: 04/21/2017] [Indexed: 02/01/2023] Open
Abstract
Vaccination is the most effective prophylactic tool against infectious diseases. Despite continued efforts to control malaria, the disease still generally represents a significant unmet medical need. Microcrystalline tyrosine (MCT) is a well described depot used in licensed allergy immunotherapy products and in clinical development. However, its proof of concept in prophylactic vaccines has only recently been explored. MCT has never been used in combination with virus-like particles (VLPs), which are considered to be one of the most potent inducers of cellular and humoral immune responses in mice and humans. In the current study we assessed the potential of MCT to serve as an adjuvant in the development of a vaccine against malaria either alone or combined with VLP using Plasmodium vivax thrombospondin-related adhesive protein (TRAP) as a target antigen. We chemically coupled PvTRAP to VLPs derived from the cucumber mosaic virus fused to a universal T-cell epitope of the tetanus toxin (CMVtt), formulated with MCT and compared the induced immune responses to PvTRAP formulated in PBS or Alum. The protective capacity of the various formulations was assessed using Plasmodium berghei expressing PvTRAP. All vaccine formulations using adjuvants and/or VLP increased humoral immunogenicity for PvTRAP compared to the antigen alone. The most proficient responder was the group of mice immunized with the vaccine formulated with PvTRAP-VLP + MCT. The VLP-based vaccine formulated in MCT also induced the strongest T cell response and conferred best protection against challenge with recombinant Plasmodium berghei. Thus, the combination of VLP with MCT may take advantage of the properties of each component and appears to be an alternative biodegradable depot adjuvant for development of novel prophylactic vaccines.
Collapse
|
11
|
Lu LL, Chung AW, Rosebrock T, Ghebremichael M, Yu WH, Grace PS, Schoen MK, Tafesse F, Martin C, Leung V, Mahan AE, Sips M, Kumar M, Tedesco J, Robinson H, Tkachenko E, Draghi M, Freedberg KJ, Streeck H, Suscovich TJ, Lauffenburger D, Restrepo BI, Day C, Fortune SM, Alter G. A Functional Role for Antibodies in Tuberculosis. Cell 2016; 167:433-443.e14. [PMID: 27667685 PMCID: PMC5526202 DOI: 10.1016/j.cell.2016.08.072] [Citation(s) in RCA: 398] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 05/04/2016] [Accepted: 08/26/2016] [Indexed: 11/18/2022]
Abstract
While a third of the world carries the burden of tuberculosis, disease control has been hindered by a lack of tools, including a rapid, point-of-care diagnostic and a protective vaccine. In many infectious diseases, antibodies (Abs) are powerful biomarkers and important immune mediators. However, in Mycobacterium tuberculosis (Mtb) infection, a discriminatory or protective role for humoral immunity remains unclear. Using an unbiased antibody profiling approach, we show that individuals with latent tuberculosis infection (Ltb) and active tuberculosis disease (Atb) have distinct Mtb-specific humoral responses, such that Ltb infection is associated with unique Ab Fc functional profiles, selective binding to FcγRIII, and distinct Ab glycosylation patterns. Moreover, compared to Abs from Atb, Abs from Ltb drove enhanced phagolysosomal maturation, inflammasome activation, and, most importantly, macrophage killing of intracellular Mtb. Combined, these data point to a potential role for Fc-mediated Ab effector functions, tuned via differential glycosylation, in Mtb control.
Collapse
Affiliation(s)
- Lenette L. Lu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA
| | - Amy W. Chung
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
- Department of Microbiology and Immunology, University of Melbourne, Doherty Institute for Infection and Immunity, Melbourne, 3000, Australia
| | - Tracy Rosebrock
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA
| | | | - Wen Han Yu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | | | | | - Fikadu Tafesse
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - Constance Martin
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA
| | - Vivian Leung
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA
| | - Alison E. Mahan
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - Magdalena Sips
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
- Department of Biomedical Molecular Biology, Ghent University, Ghent, 9000, Belgium
| | - Manu Kumar
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | | | - Hannah Robinson
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | | | - Monia Draghi
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | | | | | | | - Douglas Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA
| | - Blanca I. Restrepo
- School of Public Health, University of Texas Health Houston, Brownsville, TX, 78520, USA
| | - Cheryl Day
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, 30332, USA
- South African Tuberculosis Vaccine Initiative (SATVI) and School of Child and Adolescent Health, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Observatory 7925 South Africa
| | - Sarah M. Fortune
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| |
Collapse
|
12
|
Wen YM, Mu L, Shi Y. Immunoregulatory functions of immune complexes in vaccine and therapy. EMBO Mol Med 2016; 8:1120-1133. [PMID: 27572622 PMCID: PMC5048363 DOI: 10.15252/emmm.201606593] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 07/01/2016] [Accepted: 08/03/2016] [Indexed: 12/15/2022] Open
Abstract
Clinical and experimental preparations of IgG/soluble antigen complexes, as well as those formed following antibody therapy in vivo, are multifaceted immune regulators. These immune complexes (ICs) have been tested in humans and animal models, mostly in forms of experimental or clinical vaccination, for at least a century. With intensified research on Fcγ receptor-mediated immune modulation, as well as with immune complex-directed antigen processing, presentation, and inflammatory responses, there are renewed interests of using ICs in vaccines and immunotherapies. Currently, IC-based immune therapy has been broadly experimented in HBV and HIV viral infection control and antitumor treatments. However, mechanistic insights of IC-based treatments are relatively recent subjects of study; strong efforts are needed to establish links to connect laboratory findings with clinical practices. This review covers the history, mechanisms, and in vivo outcomes of this safe and effective therapeutic tool, with a clear aim to bridge laboratory findings with evolving clinical applications.
Collapse
Affiliation(s)
- Yu-Mei Wen
- Key Laboratory of Molecular Medical Virology, MOE/MOH, School of Basic Medical Sciences Shanghai Medical College Fudan University, Shanghai, China
| | - Libing Mu
- Center for Life Sciences, Department of Basic Medical Sciences, Institute of Immunology Tsinghua University, Beijing, China
| | - Yan Shi
- Center for Life Sciences, Department of Basic Medical Sciences, Institute of Immunology Tsinghua University, Beijing, China Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Canada
| |
Collapse
|
13
|
Konduru K, Shurtleff AC, Bradfute SB, Nakamura S, Bavari S, Kaplan G. Ebolavirus Glycoprotein Fc Fusion Protein Protects Guinea Pigs against Lethal Challenge. PLoS One 2016; 11:e0162446. [PMID: 27622456 PMCID: PMC5021345 DOI: 10.1371/journal.pone.0162446] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 08/23/2016] [Indexed: 11/26/2022] Open
Abstract
Ebola virus (EBOV), a member of the Filoviridae that can cause severe hemorrhagic fever in humans and nonhuman primates, poses a significant threat to the public health. Currently, there are no licensed vaccines or therapeutics to prevent and treat EBOV infection. Several vaccines based on the EBOV glycoprotein (GP) are under development, including vectored, virus-like particles, and protein-based subunit vaccines. We previously demonstrated that a subunit vaccine containing the extracellular domain of the Ebola ebolavirus (EBOV) GP fused to the Fc fragment of human IgG1 (EBOVgp-Fc) protected mice against EBOV lethal challenge. Here, we show that the EBOVgp-Fc vaccine formulated with QS-21, alum, or polyinosinic-polycytidylic acid-poly-L-lysine carboxymethylcellulose (poly-ICLC) adjuvants induced strong humoral immune responses in guinea pigs. The vaccinated animals developed anti-GP total antibody titers of approximately 105−106 and neutralizing antibody titers of approximately 103 as assessed by a BSL-2 neutralization assay based on vesicular stomatitis virus (VSV) pseudotypes. The poly-ICLC formulated EBOVgp-Fc vaccine protected all the guinea pigs against EBOV lethal challenge performed under BSL-4 conditions whereas the same vaccine formulated with QS-21 or alum only induced partial protection. Vaccination with a mucin-deleted EBOVgp-Fc construct formulated with QS-21 adjuvant did not have a significant effect in anti-GP antibody levels and protection against EBOV lethal challenge compared to the full-length GP construct. The bulk of the humoral response induced by the EBOVgp-Fc vaccine was directed against epitopes outside the EBOV mucin region. Our findings indicate that different adjuvants can eliciting varying levels of protection against lethal EBOV challenge in guinea pigs vaccinated with EBOVgp-Fc, and suggest that levels of total anti-GP antibodies elicit by protein-based GP subunit vaccines do not correlate with protection. Our data further support the development of Fc fusions of GP as a candidate vaccine for human use.
Collapse
Affiliation(s)
- Krishnamurthy Konduru
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States of America
- * E-mail:
| | - Amy C. Shurtleff
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, United States of America
| | - Steven B. Bradfute
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, United States of America
| | - Siham Nakamura
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States of America
| | - Sina Bavari
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, United States of America
| | - Gerardo Kaplan
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, United States of America
| |
Collapse
|
14
|
Pleass RJ, Moore SC, Stevenson L, Hviid L. Immunoglobulin M: Restrainer of Inflammation and Mediator of Immune Evasion by Plasmodium falciparum Malaria. Trends Parasitol 2015; 32:108-119. [PMID: 26597020 DOI: 10.1016/j.pt.2015.09.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 09/16/2015] [Accepted: 09/23/2015] [Indexed: 02/06/2023]
Abstract
Immunoglobulin M (IgM) is an ancient antibody class that is found in all vertebrates, with the exception of coelacanths, and is indispensable in both innate and adaptive immunity. The equally ancient human malaria parasite, Plasmodium falciparum, formed an intimate relationship with IgM with which it co-evolved. In this article, we discuss the association between IgM and human malaria parasites, building on several recent publications that implicate IgM as a crucial molecule that determines both host and parasite survival. Consequently, a better understanding of this association may lead to the development of improved intervention strategies.
Collapse
Affiliation(s)
- Richard J Pleass
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK.
| | - Shona C Moore
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK; Warwick Systems Biology Centre, Senate House, University of Warwick, Coventry, CV4 7AL, UK
| | - Liz Stevenson
- Centre for Medical Parasitology, Department of Immunology and Microbiology (ISIM), Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Lars Hviid
- Centre for Medical Parasitology, Department of Immunology and Microbiology (ISIM), Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark.
| |
Collapse
|
15
|
Grant AV, Roussilhon C, Paul R, Sakuntabhai A. The genetic control of immunity to Plasmodium infection. BMC Immunol 2015; 16:14. [PMID: 25887595 PMCID: PMC4374205 DOI: 10.1186/s12865-015-0078-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 02/20/2015] [Indexed: 12/17/2022] Open
Abstract
Background Malaria remains a major worldwide public health problem with ~207 million cases and ~627,000 deaths per year, mainly affecting children under five years of age in Africa. Recent efforts at elaborating a genetic architecture of malaria have focused on severe malaria, leading to the identification of two new genes and confirmation of previously known variants in HBB, ABO and G6PD, by exploring the whole human genome in genome-wide association (GWA) studies. Molecular pathways controlling phenotypes representing effectiveness of host immunity, notably parasitemia and IgG levels, are of particular interest given the current lack of an efficacious vaccine and the need for new treatment options. Results We propose a global causal framework of malaria phenotypes implicating progression from the initial infection with Plasmodium spp. to the development of the infection through liver and blood-stage multiplication cycles (parasitemia as a quantitative trait), to clinical malaria attack, and finally to severe malaria. Genetic polymorphism may control any of these stages, such that preceding stages act as mediators of subsequent stages. A biomarker of humoral immunity, IgG levels, can also be integrated into the framework, potentially mediating the impact of polymorphism by limiting parasitemia levels. Current knowledge of the genetic basis of parasitemia levels and IgG levels is reviewed through key examples including the hemoglobinopathies, showing that the protective effect of HBB variants on malaria clinical phenotypes may partially be mediated through parasitemia and cytophilic IgG levels. Another example is the IgG receptor FcγRIIa, encoded by FCGR2A, such that H131 homozygotes displayed higher IgG2 levels and were protective against high parasitemia and onset of malaria symptoms as shown in a causal diagram. Conclusions We thus underline the value of parasitemia and IgG levels as phenotypes in the understanding of the human genetic architecture of malaria, and the need for applying GWA approaches to these phenotypes.
Collapse
Affiliation(s)
- Audrey V Grant
- Unité de la Génétique Fonctionnelle des Maladies Infectieuses, Institut Pasteur, Paris, France. .,Centre National de la Recherche Scientifique, URA3012, Paris, France.
| | - Christian Roussilhon
- Unité de la Génétique Fonctionnelle des Maladies Infectieuses, Institut Pasteur, Paris, France. .,Centre National de la Recherche Scientifique, URA3012, Paris, France.
| | - Richard Paul
- Unité de la Génétique Fonctionnelle des Maladies Infectieuses, Institut Pasteur, Paris, France. .,Centre National de la Recherche Scientifique, URA3012, Paris, France.
| | - Anavaj Sakuntabhai
- Unité de la Génétique Fonctionnelle des Maladies Infectieuses, Institut Pasteur, Paris, France. .,Centre National de la Recherche Scientifique, URA3012, Paris, France.
| |
Collapse
|
16
|
Harris NL, Pleass R, Behnke JM. Understanding the role of antibodies in murine infections with Heligmosomoides (polygyrus) bakeri: 35 years ago, now and 35 years ahead. Parasite Immunol 2014; 36:115-24. [PMID: 23889357 DOI: 10.1111/pim.12057] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 07/24/2013] [Indexed: 12/14/2022]
Abstract
The rodent intestinal nematode H.p.bakeri has played an important role in the exploration of the host-parasite relationship of chronic nematode infections for over six decades, since the parasite was first isolated in the 1950s by Ehrenford. It soon became a popular laboratory model providing a tractable experimental system that is easy to maintain in the laboratory and far more cost-effective than other laboratory nematode-rodent model systems. Immunity to this parasite is complex, dependent on antibodies, but confounded by the parasite's potent immunosuppressive secretions that facilitate chronic survival in murine hosts. In this review, we remind readers of the state of knowledge in the 1970s, when the first volume of Parasite Immunology was published, focusing on the role of antibodies in protective immunity. We show how our understanding of the host-parasite relationship then developed over the following 35 years to date, we propose testable hypotheses for future researchers to tackle, and we speculate on how the new technologies will be applied to enable an increasingly refined understanding of the role of antibodies in host-protective immunity, and its evasion, to be achieved in the longer term.
Collapse
Affiliation(s)
- N L Harris
- Global Health Institute and Swiss Vaccine Research Institute, École Polytechnique Fédèrale de Lausanne (EPFL), Switzerland
| | | | | |
Collapse
|
17
|
Llewellyn D, de Cassan SC, Williams AR, Douglas AD, Forbes EK, Adame-Gallegos JR, Shi J, Pleass RJ, Draper SJ. Assessment of antibody-dependent respiratory burst activity from mouse neutrophils on Plasmodium yoelii malaria challenge outcome. J Leukoc Biol 2014; 95:369-82. [PMID: 24163420 PMCID: PMC3896657 DOI: 10.1189/jlb.0513274] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 09/23/2013] [Accepted: 10/01/2013] [Indexed: 01/12/2023] Open
Abstract
New tools are required to expedite the development of an effective vaccine against the blood-stage infection with the human malaria parasite Plasmodium falciparum. This work describes the assessment of the ADRB assay in a mouse model, characterizing the functional interaction between antimalarial serum antibodies and FcRs upon neutrophils. We describe a reproducible, antigen-specific assay, dependent on functional FcR signaling, and show that ADRB activity is induced equally by IgG1 and IgG2a isotypes and is modulated by blocking FcR function. However, following immunization of mice with the blood-stage vaccine candidate antigen MSP142, no measurable ADRB activity was induced against PEMS and neither was vaccine efficacy modulated against Plasmodium yoelii blood-stage challenge in γ(-/-) mice compared with WT mice. In contrast, following a primary, nonlethal P. yoelii parasite challenge, serum from vaccinated mice and nonimmunized controls showed anti-PEMS ADRB activity. Upon secondary challenge, nonimmunized γ(-/-) mice showed a reduced ability to control blood-stage parasitemia compared with immunized γ(-/-) mice; however, WT mice, depleted of their neutrophils, did not lose their ability to control infection. Thus, whereas neutrophil-induced ADRB against PEMS does not appear to play a role in protection against P. yoelii rodent malaria, induction of ADRB activity after challenge suggests that antigen targets of anti-PEMS ADRB activity remain to be established, as well as further supporting the observation that ADRB activity to P. falciparum arises following repeated natural exposure.
Collapse
Affiliation(s)
- David Llewellyn
- 1.University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Du L, Zhao G, Sun S, Zhang X, Zhou X, Guo Y, Li Y, Zhou Y, Jiang S. A critical HA1 neutralizing domain of H5N1 influenza in an optimal conformation induces strong cross-protection. PLoS One 2013; 8:e53568. [PMID: 23320093 PMCID: PMC3539987 DOI: 10.1371/journal.pone.0053568] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Accepted: 11/29/2012] [Indexed: 01/18/2023] Open
Abstract
The highly pathogenic avian influenza (HPAI) H5N1 viruses, especially the laboratory-generated H5N1 mutants, have demonstrated the potential to cross the species barrier and infect mammals and humans. Consequently, the design of an effective and safe anti-H5N1 vaccine is essential. We previously demonstrated that the full-length hemagglutinin 1 (HA1) could induce significant neutralizing antibody response and protection. Here, we intended to identify the critical neutralizing domain (CND) in an optimal conformation that can elicit strong cross-neutralizing antibodies and protection against divergent H5N1 strains. We thus constructed six recombinant proteins covering different regions of HA1 of A/Anhui/1/2005(H5N1), each of which was fused with foldon (Fd) and Fc of human IgG. We found that the critical fragment fused with Fd/Fc (HA-13–263-Fdc, H5 numbering) that could elicit the strongest neutralizing antibody response is located in the N-terminal region of HA1 (residues 13–263), which covers the receptor-binding domain (RBD, residues 112–263). We then constructed three additional recombinants fused with Fd plus His tag (HA-13–263-Fd-His), Fc only (HA-13–263-Fc), and His tag only (HA-13–263-His), respectively. We found that the HA-13–263-Fdc, which formed an oligomeric conformation, induced the strongest neutralizing antibody response and cross-protection against challenges of two tested H5N1 virus strains covering clade 1: A/VietNam/1194/2004 (VN/1194) or clade 2.3.4: A/Shenzhen/406H/06 (SZ/406H), while HA-13–263-Fc dimer and HA-13–263-Fd-His trimer elicited higher neutralizing antibody response and protection than HA-13–263-His monomer. These results suggest that the oligomeric form of the CND containing the RBD can be further developed as an effective and safe vaccine for cross-protection against divergent strains of H5N1 viruses.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/biosynthesis
- Antibodies, Neutralizing/therapeutic use
- Antibodies, Viral/biosynthesis
- Antibodies, Viral/therapeutic use
- Antigens, Viral/immunology
- Cross Reactions
- Female
- Hemagglutinin Glycoproteins, Influenza Virus/chemistry
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Humans
- Influenza A Virus, H5N1 Subtype/chemistry
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/pathogenicity
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Mice
- Mice, Inbred BALB C
- Neutralization Tests/methods
- Protein Conformation
- Protein Structure, Tertiary
- Recombinant Fusion Proteins/chemistry
- Recombinant Fusion Proteins/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, United States of America
| | - Guangyu Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Shihui Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xiujuan Zhang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, United States of America
| | - Xiaojun Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yan Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Ye Li
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, United States of America
- School of Medical Laboratory Science, Wenzhou Medical College, Wenzhou, Zhejiang, China
| | - Yusen Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
- * E-mail: (Y. Zhou); (SJ)
| | - Shibo Jiang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York, United States of America
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai, China
- * E-mail: (Y. Zhou); (SJ)
| |
Collapse
|
19
|
Forbes EK, de Cassan SC, Llewellyn D, Biswas S, Goodman AL, Cottingham MG, Long CA, Pleass RJ, Hill AVS, Hill F, Draper SJ. T cell responses induced by adenoviral vectored vaccines can be adjuvanted by fusion of antigen to the oligomerization domain of C4b-binding protein. PLoS One 2012; 7:e44943. [PMID: 22984589 PMCID: PMC3440343 DOI: 10.1371/journal.pone.0044943] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 08/14/2012] [Indexed: 11/18/2022] Open
Abstract
Viral vectored vaccines have been shown to induce both T cell and antibody responses in animals and humans. However, the induction of even higher level T cell responses may be crucial in achieving vaccine efficacy against difficult disease targets, especially in humans. Here we investigate the oligomerization domain of the α-chain of C4b-binding protein (C4 bp) as a candidate T cell "molecular adjuvant" when fused to malaria antigens expressed by human adenovirus serotype 5 (AdHu5) vectored vaccines in BALB/c mice. We demonstrate that i) C-terminal fusion of an oligomerization domain can enhance the quantity of antigen-specific CD4(+) and CD8(+) T cell responses induced in mice after only a single immunization of recombinant AdHu5, and that the T cells maintain similar functional cytokine profiles; ii) an adjuvant effect is observed for AdHu5 vectors expressing either the 42 kDa C-terminal domain of Plasmodium yoelii merozoite surface protein 1 (PyMSP1(42)) or the 83 kDa ectodomain of P. falciparum strain 3D7 apical membrane antigen 1 (PfAMA1), but not a candidate 128kDa P. falciparum MSP1 biallelic fusion antigen; iii) following two homologous immunizations of AdHu5 vaccines, antigen-specific T cell responses are further enhanced, however, in both BALB/c mice and New Zealand White rabbits no enhancement of functional antibody responses is observed; and iv) that the T cell adjuvant activity of C4 bp is not dependent on a functional Fc-receptor γ-chain in the host, but is associated with the oligomerization of small (<80 kDa) antigens expressed by recombinant AdHu5. The oligomerization domain of C4 bp can thus adjuvant T cell responses induced by AdHu5 vectors against selected antigens and its clinical utility as well as mechanism of action warrant further investigation.
Collapse
Affiliation(s)
- Emily K. Forbes
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - David Llewellyn
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Sumi Biswas
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Anna L. Goodman
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Carole A. Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases/National Institutes of Health, Rockville, Maryland, United States of America
| | | | | | | | - Simon J. Draper
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
20
|
Baschieri S. Virus Glycoproteins Tagged with the Human Fc Domain as Second Generation Vaccine Candidates. INNOVATION IN VACCINOLOGY 2012. [PMCID: PMC7122206 DOI: 10.1007/978-94-007-4543-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Traditional vaccines such as inactivated or live attenuated vaccines, are gradually giving way to more biochemically defined vaccines that are most often based on a recombinant antigen known to possess neutralizing epitopes. Such vaccines can offer improvements in speed, safety and manufacturing process but an inevitable consequence of their high degree of purification is that immunogenicity is reduced through the lack of the innate triggering molecules present in more complex preparations. Targeting recombinant vaccines to antigen presenting cells (APCs) such as dendritic cells however can improve immunogenicity by ensuring that antigen processing is as efficient as possible. Immune complexes, one of a number of routes of APC targeting, are mimicked by a recombinant approach, crystallizable fragment (Fc) fusion proteins, in which the target immunogen is linked directly to an antibody effector domain capable of interaction with receptors, FcR, on the APC cell surface. A number of virus Fc fusion proteins have been expressed in insect cells using the baculovirus expression system and shown to be efficiently produced and purified. Their use for immunization next to non-Fc tagged equivalents shows that they are powerfully immunogenic in the absence of added adjuvant and that immune stimulation is the result of the Fc-FcR interaction.
Collapse
Affiliation(s)
- Selene Baschieri
- Italian National Agency for New Technolo, Energy and Sustainable Economic Developm, ENEA, Via Anguillarese 301, Rome, 00123 Italy
| |
Collapse
|
21
|
Optimized adenovirus-antibody complexes stimulate strong cellular and humoral immune responses against an encoded antigen in naive mice and those with preexisting immunity. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 19:84-95. [PMID: 22089246 DOI: 10.1128/cvi.05319-11] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The immune response to recombinant adenoviruses is the most significant impediment to their clinical use for immunization. We test the hypothesis that specific virus-antibody combinations dictate the type of immune response generated against the adenovirus and its transgene cassette under certain physiological conditions while minimizing vector-induced toxicity. In vitro and in vivo assays were used to characterize the transduction efficiency, the T and B cell responses to the encoded transgene, and the toxicity of 1 × 10(11) adenovirus particles mixed with different concentrations of neutralizing antibodies. Complexes formed at concentrations of 500 to 0.05 times the 50% neutralizing dose (ND(50)) elicited strong virus- and transgene-specific T cell responses. The 0.05-ND(50) formulation elicited measurable anti-transgene antibodies that were similar to those of virus alone (P = 0.07). This preparation also elicited very strong transgene-specific memory T cell responses (28.6 ± 5.2% proliferation versus 7.7 ± 1.4% for virus alone). Preexisting immunity significantly reduced all responses elicited by these formulations. Although lower concentrations (0.005 and 0.0005 ND(50)) of antibody did not improve cellular and humoral responses in naïve animals, they did promote strong cellular (0.005 ND(50)) and humoral (0.0005 ND(50)) responses in mice with preexisting immunity. Some virus-antibody complexes may improve the potency of adenovirus-based vaccines in naïve individuals, while others can sway the immune response in those with preexisting immunity. Additional studies with these and other virus-antibody ratios may be useful to predict and model the type of immune responses generated against a transgene in those with different levels of exposure to adenovirus.
Collapse
|
22
|
Muxel SM, Freitas do Rosário AP, Zago CA, Castillo-Méndez SI, Sardinha LR, Rodriguez-Málaga SM, Câmara NOS, Álvarez JM, Lima MRD. The spleen CD4+ T cell response to blood-stage Plasmodium chabaudi malaria develops in two phases characterized by different properties. PLoS One 2011; 6:e22434. [PMID: 21814579 PMCID: PMC3141041 DOI: 10.1371/journal.pone.0022434] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Accepted: 06/28/2011] [Indexed: 11/19/2022] Open
Abstract
The pivotal role of spleen CD4+ T cells in the development of both malaria pathogenesis and protective immunity makes necessary a profound comprehension of the mechanisms involved in their activation and regulation during Plasmodium infection. Herein, we examined in detail the behaviour of non-conventional and conventional splenic CD4+ T cells during P. chabaudi malaria. We took advantage of the fact that a great proportion of CD4+ T cells generated in CD1d-/- mice are I-Ab-restricted (conventional cells), while their counterparts in I-Ab-/- mice are restricted by CD1d and other class IB major histocompatibility complex (MHC) molecules (non-conventional cells). We found that conventional CD4+ T cells are the main protagonists of the immune response to infection, which develops in two consecutive phases concomitant with acute and chronic parasitaemias. The early phase of the conventional CD4+ T cell response is intense and short lasting, rapidly providing large amounts of proinflammatory cytokines and helping follicular and marginal zone B cells to secrete polyclonal immunoglobulin. Both TNF-α and IFN-γ production depend mostly on conventional CD4+ T cells. IFN-γ is produced simultaneously by non-conventional and conventional CD4+ T cells. The early phase of the response finishes after a week of infection, with the elimination of a large proportion of CD4+ T cells, which then gives opportunity to the development of acquired immunity. Unexpectedly, the major contribution of CD1d-restricted CD4+ T cells occurs at the beginning of the second phase of the response, but not earlier, helping both IFN-γ and parasite-specific antibody production. We concluded that conventional CD4+ T cells have a central role from the onset of P. chabaudi malaria, acting in parallel with non-conventional CD4+ T cells as a link between innate and acquired immunity. This study contributes to the understanding of malaria immunology and opens a perspective for future studies designed to decipher the molecular mechanisms behind immune responses to Plasmodium infection.
Collapse
Affiliation(s)
- Sandra Marcia Muxel
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brazil.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Douglas AD, de Cassan SC, Dicks MDJ, Gilbert SC, Hill AVS, Draper SJ. Tailoring subunit vaccine immunogenicity: maximizing antibody and T cell responses by using combinations of adenovirus, poxvirus and protein-adjuvant vaccines against Plasmodium falciparum MSP1. Vaccine 2011; 28:7167-78. [PMID: 20937436 PMCID: PMC3404461 DOI: 10.1016/j.vaccine.2010.08.068] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2010] [Revised: 08/12/2010] [Accepted: 08/13/2010] [Indexed: 12/15/2022]
Abstract
Subunit vaccination modalities tend to induce particular immune effector responses. Viral vectors are well known for their ability to induce strong T cell responses, while protein-adjuvant vaccines have been used primarily for induction of antibody responses. Here, we demonstrate in mice using a Plasmodium falciparum merozoite surface protein 1 (PfMSP1) antigen that novel regimes combining adenovirus and poxvirus vectored vaccines with protein antigen in Montanide ISA720 adjuvant can achieve simultaneous antibody and T cell responses which equal, or in some cases surpass, the best immune responses achieved by either the viral vectors or the protein vaccine alone. Such broad responses can be achieved either using three-stage vaccination protocols, or with an equally effective two-stage protocol in which viral vectors are admixed with protein and adjuvant, and were apparent despite the use of a protein antigen that represented only a portion of the viral vector antigen. We describe further possible advantages of viral vectors in achieving consistent antibody priming, enhanced antibody avidity, and cytophilic isotype skew. These data strengthen the evidence that tailored combinations of vaccine platforms can achieve desired combinations of immune responses, and further encourage the co-administration of antibody-inducing recombinant protein vaccines with T cell- and antibody-inducing recombinant viral vectors as one strategy that may achieve protective blood-stage malaria immunity in humans.
Collapse
|
24
|
Adjuvant-free immunization with hemagglutinin-Fc fusion proteins as an approach to influenza vaccines. J Virol 2010; 85:3010-4. [PMID: 21191017 DOI: 10.1128/jvi.01241-10] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The hemagglutinins (HAs) of human H1 and H3 influenza viruses and avian H5 influenza virus were produced as recombinant fusion proteins with the human immunoglobulin Fc domain. Recombinant HA-human immunoglobulin Fc domain (HA-HuFc) proteins were secreted from baculovirus-infected insect cells as glycosylated oligomer HAs of the anticipated molecular mass, agglutinated red blood cells, were purified on protein A, and were used to immunize mice in the absence of adjuvant. Immunogenicity was demonstrated for all subtypes, with the serum samples demonstrating subtype-specific hemagglutination inhibition, epitope specificity similar to that seen with virus infection, and neutralization. HuFc-tagged HAs are potential candidates for gene-to-vaccine approaches to influenza vaccination.
Collapse
|
25
|
Goodman AL, Draper SJ. Blood-stage malaria vaccines - recent progress and future challenges. ANNALS OF TROPICAL MEDICINE AND PARASITOLOGY 2010; 104:189-211. [PMID: 20507694 DOI: 10.1179/136485910x12647085215534] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Plasmodium falciparum malaria is a major global health problem, responsible for up to 1 million deaths each year. Major efforts have been made to develop an effective vaccine against this disease, to reduce the associated morbidity and mortality. There has already been considerable progress, with the first vaccine against the pre-erythrocytic stages of P. falciparum now en route to licensure. There remains, however, a strong scientific rationale for the development of a highly effective additional vaccine component against the blood stages of the parasite, which could be deployed in conjunction with partially effective control measures against the pre-erythrocytic stages. Here, recent progress in the clinical development of blood-stage vaccines is reviewed, including methods of antigen selection, the limitations of in-vitro assays for selecting vaccines for clinical development, and the results of recently published clinical trials. This review seeks to summarize recent developments in our understanding of immunity to blood-stage parasites, as well as the relevant key advances made in vaccine technologies over the last decade. The future challenges that face this field of vaccine research are also described.
Collapse
Affiliation(s)
- A L Goodman
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK.
| | | |
Collapse
|
26
|
Gallo P, Gonçalves R, Mosser DM. The influence of IgG density and macrophage Fc (gamma) receptor cross-linking on phagocytosis and IL-10 production. Immunol Lett 2010; 133:70-7. [PMID: 20670655 DOI: 10.1016/j.imlet.2010.07.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 05/27/2010] [Accepted: 07/20/2010] [Indexed: 10/19/2022]
Abstract
We have previously demonstrated that the addition of immune complexes (IC) to stimulated macrophages could profoundly influence cytokine production. In the present work we sought to determine the density of IgG on immune complexes necessary to mediate phagocytosis, inhibit IL-12 production and induce IL-10 production from stimulated macrophages. We developed immune complexes with predictable average densities of surface-bound immunoglobulin. We show that a threshold amount of IgG was necessary to mediate attachment of IC to macrophages. At progressively higher densities of IgG, Fc receptor-mediated phagocytosis resulted in an inhibition of IL-12 production and then an induction of IL-10. The reciprocal alterations in these two cytokines occurred when as little as one optimally opsonized SRBC was bound per macrophage. Macrophage IL-10 induction by immune complexes was associated with the activation of the MAP kinase, ERK, which was progressively increased as a function of IgG density. We conclude that signal transduction through the macrophage Fcγ receptors vary as a function of signal strength. At moderate IgG densities, especially in the presence of complement, efficient phagocytosis occurs in the absence of cytokine alterations. At slightly higher IgG densities IL-12 production is shut off and eventually IL-10 induction occurs. Thus, the myriad events emanating from FcγR ligation depends on the density of immune complexes, allowing the Fc receptors to fine-tune cellular responses depending on the extent of receptor cross-linking.
Collapse
Affiliation(s)
- Paul Gallo
- Department of Cell Biology and Molecular Genetics and the Maryland Pathogen Research Institute, University of Maryland, College Park, MD 20742, USA
| | | | | |
Collapse
|
27
|
Inhibition of erythrocyte invasion and Plasmodium falciparum merozoite surface protein 1 processing by human immunoglobulin G1 (IgG1) and IgG3 antibodies. Infect Immun 2009; 77:5659-67. [PMID: 19805526 DOI: 10.1128/iai.00167-09] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antigen-specific antibodies (Abs) to the 19-kDa carboxy-terminal region of Plasmodium falciparum merozoite surface protein 1 (MSP1(19)) play an important role in protective immunity to malaria. Mouse monoclonal Abs (MAbs) 12.10 and 12.8 recognizing MSP1(19) can inhibit red cell invasion by interfering with MSP1 processing on the merozoite surface. We show here that this ability is dependent on the intact Ab since Fab and F(ab')(2) fragments derived from MAb 12.10, although capable of binding MSP1 with high affinity and competing with the intact antibody for binding to MSP1, were unable to inhibit erythrocyte invasion or MSP1 processing. The DNA sequences of the variable (V) regions of both MAbs 12.8 and 12.10 were obtained, and partial amino acid sequences of the same regions were confirmed by mass spectrometry. Human chimeric Abs constructed by using these sequences, which combine the original mouse V regions with human gamma1 and gamma3 constant regions, retain the ability to bind to both parasites and recombinant MSP1(19), and both chimeric human immunoglobulin G1s (IgG1s) were at least as good at inhibiting erythrocyte invasion as the parental murine MAbs 12.8 and 12.10. Furthermore, the human chimeric Abs of the IgG1 class (but not the corresponding human IgG3), induced significant NADPH-mediated oxidative bursts and degranulation from human neutrophils. These chimeric human Abs will enable investigators to examine the role of human Fcgamma receptors in immunity to malaria using a transgenic parasite and mouse model and may prove useful in humans for neutralizing parasites as an adjunct to antimalarial drug therapy.
Collapse
|
28
|
Pleass RJ, Behnke JM. B-cells get the T-cells but antibodies get the worms. Trends Parasitol 2009; 25:443-6. [PMID: 19734092 PMCID: PMC3115547 DOI: 10.1016/j.pt.2009.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Revised: 06/23/2009] [Accepted: 07/08/2009] [Indexed: 01/06/2023]
Abstract
Two recent papers published in Immunity and Cell Host & Microbe underline the great importance of B cells and of antibodies (Abs) in orchestrating crucial T helper cell type 2 (Th2) protective immune responses to gastrointestinal nematodes. The findings in animal models now raise major questions as to how B cells and Abs carry out these functions in humans. Here we discuss recent technological advances in humanizing animal models at the level of both Abs and their Fc-receptors, that might provide some answers.
Collapse
Affiliation(s)
- Richard J Pleass
- Institute of Genetics, and Parasite Biology and Immunogenetics Research Group, Queen's Medical Centre, University of Nottingham, NG7 2UH, UK.
| | | |
Collapse
|
29
|
Ann Stewart V, Coppel R. Issues in malaria vaccine development. Parasite Immunol 2009; 31:489-91. [PMID: 19691553 DOI: 10.1111/j.1365-3024.2009.01145.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
30
|
Pleass RJ. Platelet power: sticky problems for sticky parasites? Trends Parasitol 2009; 25:296-9. [PMID: 19539528 PMCID: PMC3116138 DOI: 10.1016/j.pt.2009.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Revised: 03/20/2009] [Accepted: 04/01/2009] [Indexed: 02/06/2023]
Abstract
Platelets might have a crucial role in the pathogenesis of both human and rodent malarias by assisting in the sequestration of infected erythrocytes within the cerebral vasculature. However, recent elegant work by McMorran et al. suggests that they are also involved in innate protection during the early stages of infection. Here, we discuss the implications of their important findings in the context of immunity to malaria.
Collapse
Affiliation(s)
- Richard J Pleass
- Institute of Genetics, School of Biology, University of Nottingham, NG7 2UH, UK.
| |
Collapse
|
31
|
Abstract
Immune complexes (ICs) are believed to play an important role in malaria pathology, and an interesting article by Mibei et al. recently published by Parasite Immunology suggests that IgG4 and IgE are particularly important. However, researchers should be aware of potential pitfalls to current assays aimed at measuring plasma ICs and correlating these to deposition in tissues.
Collapse
Affiliation(s)
- R J Pleass
- Institute of Genetics, Queen's Medical Centre, Nottingham, UK.
| |
Collapse
|