1
|
Tumwine-Downey I, Deroost K, Levy P, McLaughlin S, Hosking C, Langhorne J. Antibody-dependent immune responses elicited by blood stage-malaria infection contribute to protective immunity to the pre-erythrocytic stages. CURRENT RESEARCH IN IMMUNOLOGY 2022; 4:100054. [PMID: 36593995 PMCID: PMC9803926 DOI: 10.1016/j.crimmu.2022.100054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/21/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Advances in transcriptomics and proteomics have revealed that different life-cycle stages of the malaria parasite, Plasmodium, share antigens, thus allowing for the possibility of eliciting immunity to a parasite life-cycle stage that has not been experienced before. Using the Plasmodium chabaudi (AS strain) model of malaria in mice, we investigated how isolated exposure to blood-stage infection, bypassing a liver-stage infection, yields significant protection to sporozoite challenge resulting in lower liver parasite burdens. Antibodies are the main immune driver of this protection. Antibodies induced by blood-stage infection recognise proteins on the surface of sporozoites and can impair sporozoite gliding motility in vitro, suggesting a possible function in vivo. Furthermore, mice lacking B cells and/or secreted antibodies are not protected against a sporozoite challenge in mice that had a previous blood-stage infection. Conversely, effector CD4+ and CD8+ T cells do not seem to play a role in protection from sporozoite challenge of mice previously exposed only to the blood stages of P. chabaudi. The protective response against pre-erythrocytic stages can be induced by infections initiated by serially passaged blood-stage parasites as well as recently mosquito transmitted parasites and is effective against a different strain of P. chabaudi (CB strain), but not against another rodent malaria species, P. yoelii. The possibility to induce protective cross-stage antibodies advocates the need to consider both stage-specific and cross-stage immune responses to malaria, as natural infection elicits exposure to all life-cycle stages. Future investigation into these cross-stage antibodies allows the opportunity for candidate antigens to contribute to malaria vaccine development.
Collapse
Affiliation(s)
| | | | | | | | | | - Jean Langhorne
- Corresponding author. Malaria Immunology Laboratory, Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
2
|
Lovett B, Bilgo E, Diabate A, St Leger R. A review of progress toward field application of transgenic mosquitocidal entomopathogenic fungi. PEST MANAGEMENT SCIENCE 2019; 75:2316-2324. [PMID: 30801913 DOI: 10.1002/ps.5385] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/02/2019] [Accepted: 02/18/2019] [Indexed: 06/09/2023]
Abstract
In Africa, adult mosquito populations are primarily controlled with insecticide-impregnated bed nets and residual insecticide sprays. This coupled with widespread applications of pesticides in agriculture has led to increasing insecticide resistance in mosquito populations. We have developed multiple alternative strategies for exploiting transgenic Metarhizium spp. directed at: (i) shortening the lifespan of adult mosquitoes; (ii) reducing transmission potential of Plasmodium spp.; (iii) reducing vector competence via pre-lethal effects. The present challenge is to convert this promising strategy into a validated public health intervention by resolving outstanding issues related to the release of genetically modified organisms. © 2019 Society of Chemical Industry.
Collapse
Affiliation(s)
- Brian Lovett
- Department of Entomology, University of Maryland, College Park, MD, USA
| | - Etienne Bilgo
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Abdoulaye Diabate
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Raymond St Leger
- Department of Entomology, University of Maryland, College Park, MD, USA
| |
Collapse
|
3
|
Kingston NJ, Kurtovic L, Walsh R, Joe C, Lovrecz G, Locarnini S, Beeson JG, Netter HJ. Hepatitis B virus-like particles expressing Plasmodium falciparum epitopes induce complement-fixing antibodies against the circumsporozoite protein. Vaccine 2019; 37:1674-1684. [PMID: 30773400 DOI: 10.1016/j.vaccine.2019.01.056] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/11/2019] [Accepted: 01/18/2019] [Indexed: 12/21/2022]
Abstract
The repetitive structure of compact virus-like particles (VLPs) provides high density displays of antigenic sequences, which trigger key parts of the immune system. The hepatitis B virus (HBV) and human papilloma virus (HPV) vaccines exploit the assembly competence of structural proteins, which are the effective immunogenic components of the prophylactic HBV and HPV vaccines, respectively. To optimize vaccine designs and to promote immune responses against protective epitopes, the "Asp-Ala-Asp-Pro" (NANP)-repeat from the Plasmodium falciparum circumsporozoite protein (CSP) was expressed within the exposed, main antigenic site of the small HBV envelope protein (HBsAgS); this differs from the RTS,S vaccine, in which CSP epitopes are fused to the N-terminus of HBsAgS. The chimeric HBsAgS proteins are assembly competent, produce VLPs, and provide a high antigenic density of the NANP repeat sequence. Chimeric VLPs with four or nine NANP-repeats (NANP4 and NANP9, respectively) were expressed in mammalian cells, the HBsAgS- and CSP-specific antigenicity of the VLPs was determined, and the immunogenicity of the VLPs assessed in relation to the induction of anti-HBsAgS and anti-CSP antibody responses. The chimeric VLPs induced high anti-CSP titres in BALB/c mice independent of the number of the NANP repeats. However, the number of NANP repeats influenced the activity of vaccine-induced antibodies measured by complement fixation to CSP, one of the proposed effector mechanisms for Plasmodium neutralization in vivo. Sera from mice immunized with VLPs containing nine NANP repeats performed better in the complement fixation assay than the group with four NANP repeats. The effect of the epitope-specific density on the antibody quality may instruct VLP platform designs to optimize immunological outcomes and vaccine efficacy.
Collapse
Affiliation(s)
- Natalie J Kingston
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia; School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Liriye Kurtovic
- Burnet Institute, Commercial Road, Melbourne, Victoria 3004, Australia; Department of Immunology and Pathology, Monash University, Melbourne, Victoria 2004, Australia
| | - Renae Walsh
- Victorian Infectious Diseases Reference Laboratory (VIDRL), Melbourne Health, The Peter Doherty Institute, Melbourne, Victoria 3000, Australia
| | - Carina Joe
- Royal Melbourne Institute of Technology (RMIT) University, School of Science, Melbourne, Victoria 3001, Australia; Commonwealth Scientific and Industrial Research Organisation, Clayton, Victoria 3169, Australia
| | - George Lovrecz
- Commonwealth Scientific and Industrial Research Organisation, Clayton, Victoria 3169, Australia
| | - Stephen Locarnini
- Victorian Infectious Diseases Reference Laboratory (VIDRL), Melbourne Health, The Peter Doherty Institute, Melbourne, Victoria 3000, Australia
| | - James G Beeson
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia; Burnet Institute, Commercial Road, Melbourne, Victoria 3004, Australia; Department of Immunology and Pathology, Monash University, Melbourne, Victoria 2004, Australia; Department of Medicine, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Hans J Netter
- Victorian Infectious Diseases Reference Laboratory (VIDRL), Melbourne Health, The Peter Doherty Institute, Melbourne, Victoria 3000, Australia; Royal Melbourne Institute of Technology (RMIT) University, School of Science, Melbourne, Victoria 3001, Australia.
| |
Collapse
|
4
|
Fisher CR, Sutton HJ, Kaczmarski JA, McNamara HA, Clifton B, Mitchell J, Cai Y, Dups JN, D'Arcy NJ, Singh M, Chuah A, Peat TS, Jackson CJ, Cockburn IA. T-dependent B cell responses to Plasmodium induce antibodies that form a high-avidity multivalent complex with the circumsporozoite protein. PLoS Pathog 2017; 13:e1006469. [PMID: 28759640 PMCID: PMC5552345 DOI: 10.1371/journal.ppat.1006469] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 08/10/2017] [Accepted: 06/13/2017] [Indexed: 11/18/2022] Open
Abstract
The repeat region of the Plasmodium falciparum circumsporozoite protein (CSP) is a major vaccine antigen because it can be targeted by parasite neutralizing antibodies; however, little is known about this interaction. We used isothermal titration calorimetry, X-ray crystallography and mutagenesis-validated modeling to analyze the binding of a murine neutralizing antibody to Plasmodium falciparum CSP. Strikingly, we found that the repeat region of CSP is bound by multiple antibodies. This repeating pattern allows multiple weak interactions of single FAB domains to accumulate and yield a complex with a dissociation constant in the low nM range. Because the CSP protein can potentially cross-link multiple B cell receptors (BCRs) we hypothesized that the B cell response might be T cell independent. However, while there was a modest response in mice deficient in T cell help, the bulk of the response was T cell dependent. By sequencing the BCRs of CSP-repeat specific B cells in inbred mice we found that these cells underwent somatic hypermutation and affinity maturation indicative of a T-dependent response. Last, we found that the BCR repertoire of responding B cells was limited suggesting that the structural simplicity of the repeat may limit the breadth of the immune response. Vaccines aim to protect by inducing the immune system to make molecules called antibodies that can recognize molecules on the surface of invading pathogens. In the case of malaria, our most advanced vaccine candidates aim to promote the production of antibodies that recognize the circumsporozoite protein (CSP) molecule on the surface of the invasive parasite stage called the sporozoite. In this report we use X-ray crystallography to determine the structure of CSP-binding antibodies at the atomic level. We use other techniques such as isothermal titration calorimetry and structural modeling to examine how this antibody interacts with the CSP molecule. Strikingly, we found that each CSP molecule could bind 6 antibodies. This finding has implications for the immune response and may explain why high titers of antibody are needed for protection. Moreover, because the structure of the CSP repeat is quite simple we determined that the number of different kinds of antibodies that could bind this molecule are quite small. However a high avidity interaction between those antibodies and CSP can result from a process called affinity maturation that allows the body to learn how to make improved antibodies specific for pathogen molecules. These data show that while it is challenging for the immune system to recognize and neutralize CSP, it should be possible to generate viable vaccines targeting this molecule.
Collapse
Affiliation(s)
- Camilla R. Fisher
- Research School of Chemistry, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Henry J. Sutton
- John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Joe A. Kaczmarski
- Research School of Chemistry, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Hayley A. McNamara
- John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Ben Clifton
- Research School of Chemistry, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Joshua Mitchell
- Research School of Chemistry, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Yeping Cai
- John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Johanna N. Dups
- John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Nicholas J. D'Arcy
- John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Mandeep Singh
- John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Aaron Chuah
- John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Thomas S. Peat
- CSIRO Biomedical Manufacturing Program, Parkville, Victoria, Australia
| | - Colin J. Jackson
- Research School of Chemistry, The Australian National University, Canberra, Australian Capital Territory, Australia
- * E-mail: (CJJ); (IAC)
| | - Ian A. Cockburn
- John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
- * E-mail: (CJJ); (IAC)
| |
Collapse
|
5
|
Zhao H, Lovett B, Fang W. Genetically Engineering Entomopathogenic Fungi. ADVANCES IN GENETICS 2016; 94:137-63. [PMID: 27131325 DOI: 10.1016/bs.adgen.2015.11.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Entomopathogenic fungi have been developed as environmentally friendly alternatives to chemical insecticides in biocontrol programs for agricultural pests and vectors of disease. However, mycoinsecticides currently have a small market share due to low virulence and inconsistencies in their performance. Genetic engineering has made it possible to significantly improve the virulence of fungi and their tolerance to adverse conditions. Virulence enhancement has been achieved by engineering fungi to express insect proteins and insecticidal proteins/peptides from insect predators and other insect pathogens, or by overexpressing the pathogen's own genes. Importantly, protein engineering can be used to mix and match functional domains from diverse genes sourced from entomopathogenic fungi and other organisms, producing insecticidal proteins with novel characteristics. Fungal tolerance to abiotic stresses, especially UV radiation, has been greatly improved by introducing into entomopathogens a photoreactivation system from an archaean and pigment synthesis pathways from nonentomopathogenic fungi. Conversely, gene knockout strategies have produced strains with reduced ecological fitness as recipients for genetic engineering to improve virulence; the resulting strains are hypervirulent, but will not persist in the environment. Coupled with their natural insect specificity, safety concerns can also be mitigated by using safe effector proteins with selection marker genes removed after transformation. With the increasing public concern over the continued use of synthetic chemical insecticides and growing public acceptance of genetically modified organisms, new types of biological insecticides produced by genetic engineering offer a range of environmentally friendly options for cost-effective control of insect pests.
Collapse
Affiliation(s)
- H Zhao
- Zhejiang University, Hangzhou, Zhejiang, China
| | - B Lovett
- University of Maryland, College Park, MD, United States
| | - W Fang
- Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Maskus DJ, Bethke S, Seidel M, Kapelski S, Addai-Mensah O, Boes A, Edgü G, Spiegel H, Reimann A, Fischer R, Barth S, Klockenbring T, Fendel R. Isolation, production and characterization of fully human monoclonal antibodies directed to Plasmodium falciparum MSP10. Malar J 2015; 14:276. [PMID: 26174014 PMCID: PMC4502606 DOI: 10.1186/s12936-015-0797-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 07/07/2015] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Semi-immunity against the malaria parasite is defined by a protection against clinical episodes of malaria and is partially mediated by a repertoire of inhibitory antibodies directed against the blood stage of Plasmodium falciparum, in particular against surface proteins of merozoites, the invasive form of the parasite. Such antibodies may be used for preventive or therapeutic treatment of P. falciparum malaria. Here, the isolation and characterization of novel human monoclonal antibodies (humAbs) for such applications is described. METHODS B lymphocytes had been selected by flow cytometry for specificity against merozoite surface proteins, including the merozoite surface protein 10 (MSP10). After Epstein-Barr virus (EBV) transformation and identification of promising resulting lymphoblastoid cell lines (LCLs), human immunoglobulin heavy and light chain variable regions (Vh or Vl regions) were secured, cloned into plant expression vectors and transiently produced in Nicotiana benthamiana in the context of human full-size IgG1:κ. The specificity and the affinity of the generated antibodies were assessed by ELISA, dotblot and surface plasmon resonance (SPR) spectroscopy. The growth inhibitory activity was evaluated based on growth inhibition assays (GIAs) using the parasite strain 3D7A. RESULTS Supernatants from two LCLs, 5E8 and 5F6, showed reactivity against the second (5E8) or first (5F6) epidermal growth factor (EGF)-like domain of MSP10. The isolated V regions were recombinantly expressed in their natural pairing as well as in combination with each other. The resulting recombinant humAbs showed affinities of 9.27 × 10(-7) M [humAb10.1 (H5F6:κ5E8)], 5.46 × 10(-9) M [humAb10.2 (H5F6:κ5F6)] and 4.34 × 10(-9) M [humAb10.3 (H5E8:κ5E8)]. In GIAs, these antibodies exhibited EC50 values of 4.1 mg/ml [95% confidence interval (CI) 2.6-6.6 mg/ml], 6.9 mg/ml (CI 5.5-8.6 mg/ml) and 9.5 mg/ml (CI 5.5-16.4 mg/ml), respectively. CONCLUSION This report describes a platform for the isolation of human antibodies from semi-immune blood donors by EBV transformation and their subsequent characterization after transient expression in plants. To our knowledge, the presented antibodies are the first humAbs directed against P. falciparum MSP10 to be described. They recognize the EGF-like folds of MSP10 and bind these with high affinity. Moreover, these antibodies inhibit P. falciparum 3D7A growth in vitro.
Collapse
Affiliation(s)
- Dominika J Maskus
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany.
- Institute for Molecular Biotechnology, RWTH Aachen University, Aachen, Germany.
| | - Susanne Bethke
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany.
| | - Melanie Seidel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany.
| | - Stephanie Kapelski
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany.
- Institute for Molecular Biotechnology, RWTH Aachen University, Aachen, Germany.
| | - Otchere Addai-Mensah
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany.
- Institute for Molecular Biotechnology, RWTH Aachen University, Aachen, Germany.
- Faculty of Allied Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana.
| | - Alexander Boes
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany.
| | - Güven Edgü
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany.
| | - Holger Spiegel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany.
| | - Andreas Reimann
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany.
| | - Rainer Fischer
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany.
- Institute for Molecular Biotechnology, RWTH Aachen University, Aachen, Germany.
| | - Stefan Barth
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany.
- Department of Experimental Medicine and Immunotherapy, Institute for Applied Medical Engineering at RWTH Aachen University and Hospital, Aachen, Germany.
| | - Torsten Klockenbring
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany.
| | - Rolf Fendel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany.
- Institute for Molecular Biotechnology, RWTH Aachen University, Aachen, Germany.
- Department of Experimental Medicine and Immunotherapy, Institute for Applied Medical Engineering at RWTH Aachen University and Hospital, Aachen, Germany.
| |
Collapse
|
7
|
Fang W, Azimzadeh P, St. Leger RJ. Strain improvement of fungal insecticides for controlling insect pests and vector-borne diseases. Curr Opin Microbiol 2012; 15:232-8. [DOI: 10.1016/j.mib.2011.12.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Accepted: 12/22/2011] [Indexed: 01/28/2023]
|
8
|
Ribosome display of combinatorial antibody libraries derived from mice immunized with heat-killed Xylella fastidiosa and the selection of MopB-specific single-chain antibodies. Appl Environ Microbiol 2012; 78:2638-47. [PMID: 22327580 DOI: 10.1128/aem.07807-11] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Pierce's disease is a devastating lethal disease of Vitus vinifera grapevines caused by the bacterium Xylella fastidiosa. There is no cure for Pierce's disease, and control is achieved predominantly by suppressing transmission of the glassy-winged sharpshooter insect vector. We present a simple robust approach for the generation of panels of recombinant single-chain antibodies against the surface-exposed elements of X. fastidiosa that may have potential use in diagnosis and/or disease transmission blocking studies. In vitro combinatorial antibody ribosome display libraries were assembled from immunoglobulin transcripts rescued from the spleens of mice immunized with heat-killed X. fastidiosa. The libraries were used in a single round of selection against an outer membrane protein, MopB, resulting in the isolation of a panel of recombinant antibodies. The potential use of selected anti-MopB antibodies was demonstrated by the successful application of the 4XfMopB3 antibody in an enzyme-linked immunosorbent assay (ELISA), a Western blot assay, and an immunofluorescence assay (IFA). These immortalized in vitro recombinant single-chain antibody libraries generated against heat-killed X. fastidiosa are a resource for the Pierce's disease research community that may be readily accessed for the isolation of antibodies against a plethora of X. fastidiosa surface-exposed antigenic molecules.
Collapse
|
9
|
Identification of a specific region of Plasmodium falciparum EBL-1 that binds to host receptor glycophorin B and inhibits merozoite invasion in human red blood cells. Mol Biochem Parasitol 2012; 183:23-31. [PMID: 22273481 DOI: 10.1016/j.molbiopara.2012.01.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 01/05/2012] [Accepted: 01/09/2012] [Indexed: 11/22/2022]
Abstract
The malaria parasite Plasmodium falciparum invades human erythrocytes through multiple pathways utilizing several ligand-receptor interactions. These interactions are broadly classified in two groups according to their dependency on sialic acid residues. Here, we focus on the sialic acid-dependent pathway by using purified glycophorins and red blood cells (RBCs) to screen a cDNA phage display library derived from P. falciparum FCR3 strain, a sialic acid-dependent strain. This screen identified several parasite proteins including the erythrocyte-binding ligand-1, EBL-1. The phage cDNA insert encoded the 69-amino acid peptide, termed F2i, which is located within the F2 region of the DBL domain, designated here as D2, of EBL-1. Recombinant D2 and F2i polypeptides bound to purified glycophorins and RBCs, and the F2i peptide was found to interfere with binding of D2 domain to its receptor. Both D2 and F2i polypeptides bound to trypsin-treated but not neuraminidase or chymotrypsin-treated erythrocytes, consistent with known glycophorin B resistance to trypsin, and neither the D2 nor F2i polypeptide bound to glycophorin B-deficient erythrocytes. Importantly, purified D2 and F2i polypeptides partially inhibited merozoite reinvasion in human erythrocytes. Our results show that the host erythrocyte receptor glycophorin B directly interacts with the DBL domain of parasite EBL-1, and the core binding site is contained within the 69 amino acid F2i region (residues 601-669) of the DBL domain. Together, these findings suggest that a recombinant F2i peptide with stabilized structure could provide a protective function at blood stage infection and represents a valuable addition to a multi-subunit vaccine against malaria.
Collapse
|
10
|
Markiv A, Beatson R, Burchell J, Durvasula RV, Kang AS. Expression of recombinant multi-coloured fluorescent antibodies in gor -/trxB- E. coli cytoplasm. BMC Biotechnol 2011; 11:117. [PMID: 22129156 PMCID: PMC3280946 DOI: 10.1186/1472-6750-11-117] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 11/30/2011] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Antibody-fluorophore conjugates are invaluable reagents used in contemporary molecular cell biology for imaging, cell sorting and tracking intracellular events. However they suffer in some cases from batch to batch variation, partial loss of binding and susceptibility to photo-bleaching. In theory, these issues can all be addressed by using recombinant antibody fused directly to genetically encoded fluorescent reporters. However, single-chain fragment variable domains linked by long flexible linkers are themselves prone to disassociation and aggregation, and in some cases with isoelectric points incompatible with use in physiologically relevant milieu. Here we describe a general approach that permits fully functional intracellular production of a range of coloured fluorescent recombinant antibodies with optimally orientated VH/VL interfaces and isoelectric points compatible for use in physiological solutions at pH 7.4 with a binding site to fluorophore stoichiometry of 1:1. RESULTS Here we report the design, assembly, intracellular bacterial production and purification of a panel of novel antibody fluorescent protein fusion constructs. The insertion of monomeric fluorescent protein derived from either Discosoma or Aequorea in-between the variable regions of anti-p185HER2-ECD antibody 4D5-8 resulted in optimal VH/VL interface interactions to create soluble coloured antibodies each with a single binding site, with isoelectric points of 6.5- 6. The fluorescent antibodies used in cell staining studies with SK-BR-3 cells retained the fluorophore properties and antibody specificity functions, whereas the conventional 4D5-8 single chain antibody with a (Gly4Ser)3 linker precipitated at physiological pH 7.4. CONCLUSIONS This modular monomeric recombinant fluorescent antibody platform may be used to create a range of recombinant coloured antibody molecules for quantitative in situ, in vivo and ex vivo imaging, cell sorting and cell trafficking studies. Assembling the single chain antibody with monomeric fluorescent protein linker facilitates optimal variable domain pairing and alters the isoelectric point of the recombinant 4D5-8 protein conferring solubility at physiological pH 7.4. The efficient intracellular expression of these functional molecules opens up the possibility of developing an alternative approach for tagging intracellular targets with fluorescent proteins for a range of molecular cell biology imaging studies.
Collapse
Affiliation(s)
- Anatoliy Markiv
- School of Life Sciences, University of Westminster, 115 New Cavendish St, London, W1W 6UW, UK
| | | | | | | | | |
Collapse
|
11
|
Herrera S, Fernández OL, Vera O, Cárdenas W, Ramírez O, Palacios R, Chen-Mok M, Corradin G, Arévalo-Herrera M. Phase I safety and immunogenicity trial of Plasmodium vivax CS derived long synthetic peptides adjuvanted with montanide ISA 720 or montanide ISA 51. Am J Trop Med Hyg 2011; 84:12-20. [PMID: 21292873 DOI: 10.4269/ajtmh.2011.09-0516] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
We assessed the safety, tolerability, and immunogenicity of a mixture of three synthetic peptides derived from the Plasmodium vivax circumsporozoite protein formulated in Montanide ISA 720 or Montanide ISA 51. Forty healthy malaria-naive volunteers were allocated to five experimental groups (A-E): four groups (A-D) were immunized intramuscularly with 50 and 100 μg/dose injections of a mixture of N, R, and C peptides formulated in the two different adjuvants at 0, 2, and 4 months and one group was administered placebo. Vaccines were immunogenic, safe, well tolerated, and no serious adverse events related to the vaccine occurred. Seroconversion occurred in > 90% of the vaccines and antibodies recognized the sporozoite protein on immunofluorescent antibody test. Vaccines in Montanide ISA 51 showed a higher sporozoite protein recognition and interferon production. Results encourage further testing of the vaccine protective efficacy.
Collapse
Affiliation(s)
- Sócrates Herrera
- Instituto de Inmunología, Facultad de Salud, Universidad del Valle, Cali, Colombia.
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Fang W, Vega-Rodríguez J, Ghosh AK, Jacobs-Lorena M, Kang A, St. Leger RJ. Development of transgenic fungi that kill human malaria parasites in mosquitoes. Science 2011; 331:1074-7. [PMID: 21350178 PMCID: PMC4153607 DOI: 10.1126/science.1199115] [Citation(s) in RCA: 177] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Metarhizium anisopliae infects mosquitoes through the cuticle and proliferates in the hemolymph. To allow M. anisopliae to combat malaria in mosquitoes with advanced malaria infections, we produced recombinant strains expressing molecules that target sporozoites as they travel through the hemolymph to the salivary glands. Eleven days after a Plasmodium-infected blood meal, mosquitoes were treated with M. anisopliae expressing salivary gland and midgut peptide 1 (SM1), which blocks attachment of sporozoites to salivary glands; a single-chain antibody that agglutinates sporozoites; or scorpine, which is an antimicrobial toxin. These reduced sporozoite counts by 71%, 85%, and 90%, respectively. M. anisopliae expressing scorpine and an [SM1](8):scorpine fusion protein reduced sporozoite counts by 98%, suggesting that Metarhizium-mediated inhibition of Plasmodium development could be a powerful weapon for combating malaria.
Collapse
Affiliation(s)
- Weiguo Fang
- Department of Entomology, University of Maryland, 4112 Plant Sciences Building, College Park, MD 20742, USA
| | - Joel Vega-Rodríguez
- Malaria Research Institute, Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, 615 North Wolfe Street, E5132, Baltimore, MD 21205, USA
| | - Anil K. Ghosh
- Malaria Research Institute, Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, 615 North Wolfe Street, E5132, Baltimore, MD 21205, USA
| | - Marcelo Jacobs-Lorena
- Malaria Research Institute, Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, 615 North Wolfe Street, E5132, Baltimore, MD 21205, USA
| | - Angray Kang
- Antibody Technology Group, Department of Molecular and Applied Biosciences, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK
| | - Raymond J. St. Leger
- Department of Entomology, University of Maryland, 4112 Plant Sciences Building, College Park, MD 20742, USA
| |
Collapse
|
13
|
Nguyen TV, Sacci JB, de la Vega P, John CC, James AA, Kang AS. Characterization of immunoglobulin G antibodies to Plasmodium falciparum sporozoite surface antigen MB2 in malaria exposed individuals. Malar J 2009; 8:235. [PMID: 19852802 PMCID: PMC2772840 DOI: 10.1186/1475-2875-8-235] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2009] [Accepted: 10/23/2009] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND MB2 protein is a sporozoite surface antigen on the human malaria parasite Plasmodium falciparum. MB2 was identified by screening a P. falciparum sporozoite cDNA expression library using immune sera from a protected donor immunized via the bites of P. falciparum-infected irradiated mosquitoes. It is not known whether natural exposure to P. falciparum also induces the anti-MB2 response and if this response differs from that in protected individuals immunized via the bites of P. falciparum infected irradiated mosquitoes. The anti-MB2 antibody response may be part of a robust protective response against the sporozoite. METHODS Fragments of polypeptide regions of MB2 were constructed as recombinant fusions sandwiched between glutathione S-transferase and a hexa histidine tag for bacterial expression. The hexa histidine tag affinity purified proteins were used to immunize rabbits and the polyclonal sera evaluated in an in vitro inhibition of sporozoite invasion assay. The proteins were also used in immunoblots with sera from a limited number of donors immunized via the bites of P. falciparum infected irradiated mosquitoes and plasma and serum obtained from naturally exposed individuals in Kenya. RESULTS Rabbit polyclonal antibodies targeting the non-repeat region of the basic domain of MB2 inhibited sporozoites entry into HepG2-A16 cells in vitro. Analysis of serum from five human volunteers that were immunized via the bites of P. falciparum infected irradiated mosquitoes that developed immunity and were completely protected against subsequent challenge with non-irradiated parasite also had detectable levels of antibody against MB2 basic domain. In contrast, in three volunteers not protected, anti-MB2 antibodies were below the level of detection. Sera from protected volunteers preferentially recognized a non-repeat region of the basic domain of MB2, whereas plasma from naturally-infected individuals also had antibodies that recognize regions of MB2 that contain a repeat motif in immunoblots. Sequence analysis of eleven field isolates and four laboratory strains showed that these antigenic regions of the basic domain of the MB2 gene are highly conserved in parasites obtained from different parts of the world. Moreover, anti-MB2 antibodies also were detected in the plasma of 83% of the individuals living in a malaria endemic area of Kenya (n = 41). CONCLUSION A preliminary analysis of the human humoral response against MB2 indicates that it may be an additional highly conserved target for immune intervention at the pre-erythrocytic stage of P. falciparum life cycle.
Collapse
Affiliation(s)
- Thanh V Nguyen
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
- NeoGenomics California, 6 Morgan, Suite 150, Irvine, CA 92618, USA
| | - John B Sacci
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Room 324 660 W Redwood Street, Baltimore, MD 21201, USA
| | - Patricia de la Vega
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Room 324 660 W Redwood Street, Baltimore, MD 21201, USA
- Department of Cell Mediated Immunity, Division of Malaria Vaccine Development, US Military Malaria Vaccine Program, Walter Reed Army Institute of Research, USA
| | - Chandy C John
- Global Pediatrics Program and Division of Pediatric Infectious Diseases, University of MN Medical School, 420 Delaware Street, SE, MMC #296, 850-Mayo, Minneapolis, MN 55455, USA
| | - Anthony A James
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
- Department of Microbiology and Molecular Genetics, University of California, Irvine, CA 92697-3900, USA
| | - Angray S Kang
- The School of Life Sciences, Department of Molecular and Applied Biosciences, University of Westminster, 115 New Cavendish Street, London, W1W 6UW, UK
| |
Collapse
|
14
|
Phage display: a useful tool for malaria research? Trends Parasitol 2007; 24:18-23. [PMID: 18037345 DOI: 10.1016/j.pt.2007.09.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2007] [Revised: 08/24/2007] [Accepted: 09/21/2007] [Indexed: 11/15/2022]
Abstract
Defining the molecular intricacies of malaria pathogenesis is a vital area of medical and scientific research. Sophisticated methods have been developed to identify and characterise host-parasite interactions that are important in infection. Phage display involves the combinatorial display of proteins or peptides on the surface of bacteriophage. The technology provides an invaluable tool for screening diverse libraries for polypeptides that have a high affinity for a given target. Phage display in malaria research has proven successful, not only in mapping the protein-protein interactions that are important in Plasmodium biology, but also in the identification of molecules that might be exploited in the design of therapeutic agents or vaccines.
Collapse
|
15
|
Yadava A, Sattabongkot J, Washington MA, Ware LA, Majam V, Zheng H, Kumar S, Ockenhouse CF. A novel chimeric Plasmodium vivax circumsporozoite protein induces biologically functional antibodies that recognize both VK210 and VK247 sporozoites. Infect Immun 2006; 75:1177-85. [PMID: 17158893 PMCID: PMC1828583 DOI: 10.1128/iai.01667-06] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A successful vaccine against Plasmodium vivax malaria would significantly improve the health and quality of the lives of more than 1 billion people around the world. A subunit vaccine is the only option in the absence of long-term culture of P. vivax parasites. The circumsporozoite protein that covers the surface of Plasmodium sporozoites is one of the best-studied malarial antigens and the most promising vaccine in clinical trials. We report here the development of a novel "immunologically optimal" recombinant vaccine expressed in Escherichia coli that encodes a chimeric CS protein encompassing repeats from the two major alleles, VK210 and VK247. This molecule is widely recognized by sera from patients naturally exposed to P. vivax infection and induces a highly potent immune response in genetically disparate strains of mice. Antibodies from immunized animals recognize both VK210 and VK247 sporozoites. Furthermore, these antibodies appear to be protective in nature since they cause the agglutination of live sporozoites, an in vitro surrogate of sporozoite infectivity. These results strongly suggest that recombinant CS is biologically active and highly immunogenic across major histocompatibility complex strains and raises the prospect that in humans this vaccine may induce protective immune responses.
Collapse
Affiliation(s)
- Anjali Yadava
- Division of Malaria Vaccine Development, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Garcia JE, Puentes A, Patarroyo ME. Developmental biology of sporozoite-host interactions in Plasmodium falciparum malaria: implications for vaccine design. Clin Microbiol Rev 2006; 19:686-707. [PMID: 17041140 PMCID: PMC1592691 DOI: 10.1128/cmr.00063-05] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Plasmodium falciparum sporozoite infects different types of cells in a mosquito's salivary glands and human epithelial and Kuppfer cells and hepatocytes. These become differentiated later on, transforming themselves into the invasive red blood cell form, the merozoite. The ability of sporozoites to interact with different types of cells requires a wide variety of mechanisms allowing them to survive in both hosts: mobility, receptor-ligand interactions with different cellular receptors, and transformation and development into other invasive parasite forms, which are vitally important for parasite survival. Sporozoite complexity is reflected in the large quantity of proteins that can be expressed. Some of them have been extensively studied, such as CSP, TRAP, STARP, LSA-1, LSA-3, SALSA, SPECT1, SPECT2, MAEBL, and SPATR, due to their importance in infection and their potential use as vaccines. Our work has been focused on the search for the molecular mechanisms of parasite-host cellular receptor-ligand interactions by identifying amino acid sequences and the critical binding residues from these proteins relevant to parasite invasion. Once such sequences have been identified, it will be possible to modify them to induce a strong immune response against P. falciparum in the experimental Aotus monkey model. This all leads towards developing multistage, multicomponent, subunit-based vaccines that will be effective in eradicating or controlling malaria caused by P. falciparum.
Collapse
Affiliation(s)
- Javier E Garcia
- Fundacion Instituto de Immunología de Colombia, Carrera 50 #26-00, Bogotá, Colombia
| | | | | |
Collapse
|
17
|
Stratmann T, Kang AS. Cognate peptide-receptor ligand mapping by directed phage display. Proteome Sci 2005; 3:7. [PMID: 15963231 PMCID: PMC1183247 DOI: 10.1186/1477-5956-3-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2004] [Accepted: 06/17/2005] [Indexed: 12/01/2022] Open
Abstract
Background A rapid phage display method for the elucidation of cognate peptide specific ligand for receptors is described. The approach may be readily integrated into the interface of genomic and proteomic studies to identify biologically relevant ligands. Methods A gene fragment library from influenza coat protein haemagglutinin (HA) gene was constructed by treating HA cDNA with DNAse I to create 50 – 100 bp fragments. These fragments were cloned into plasmid pORFES IV and in-frame inserts were selected. These in-frame fragment inserts were subsequently cloned into a filamentous phage display vector JC-M13-88 for surface display as fusions to a synthetic copy of gene VIII. Two well characterized antibodies, mAb 12CA5 and pAb 07431, directed against distinct known regions of HA were used to pan the library. Results Two linear epitopes, HA peptide 112 – 126 and 162–173, recognized by mAb 12CA5 and pAb 07431, respectively, were identified as the cognate epitopes. Conclusion This approach is a useful alternative to conventional methods such as screening of overlapping synthetic peptide libraries or gene fragment expression libraries when searching for precise peptide protein interactions, and may be applied to functional proteomics.
Collapse
Affiliation(s)
- Thomas Stratmann
- Department of Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
- Universidad de Barcelona, Departamento de Fisiologia, Diagonal 645, 3°, 08028 Barcelona, Spain
| | - Angray S Kang
- Department of Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|