1
|
Angage D, Chmielewski J, Maddumage JC, Hesping E, Caiazzo S, Lai KH, Yeoh LM, Menassa J, Opi DH, Cairns C, Puthalakath H, Beeson JG, Kvansakul M, Boddey JA, Wilson DW, Anders RF, Foley M. A broadly cross-reactive i-body to AMA1 potently inhibits blood and liver stages of Plasmodium parasites. Nat Commun 2024; 15:7206. [PMID: 39174515 PMCID: PMC11341838 DOI: 10.1038/s41467-024-50770-7] [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: 12/12/2023] [Accepted: 07/19/2024] [Indexed: 08/24/2024] Open
Abstract
Apical membrane antigen-1 (AMA1) is a conserved malarial vaccine candidate essential for the formation of tight junctions with the rhoptry neck protein (RON) complex, enabling Plasmodium parasites to invade human erythrocytes, hepatocytes, and mosquito salivary glands. Despite its critical role, extensive surface polymorphisms in AMA1 have led to strain-specific protection, limiting the success of AMA1-based interventions beyond initial clinical trials. Here, we identify an i-body, a humanised single-domain antibody-like molecule that recognises a conserved pan-species conformational epitope in AMA1 with low nanomolar affinity and inhibits the binding of the RON2 ligand to AMA1. Structural characterisation indicates that the WD34 i-body epitope spans the centre of the conserved hydrophobic cleft in AMA1, where interacting residues are highly conserved among all Plasmodium species. Furthermore, we show that WD34 inhibits merozoite invasion of erythrocytes by multiple Plasmodium species and hepatocyte invasion by P. falciparum sporozoites. Despite a short half-life in mouse serum, we demonstrate that WD34 transiently suppressed P. berghei infections in female BALB/c mice. Our work describes the first pan-species AMA1 biologic with inhibitory activity against multiple life-cycle stages of Plasmodium. With improved pharmacokinetic characteristics, WD34 could be a potential immunotherapy against multiple species of Plasmodium.
Collapse
Affiliation(s)
- Dimuthu Angage
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Sciences, La Trobe University, Victoria, 3086, Australia
| | - Jill Chmielewski
- Research Centre for Infectious Diseases, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Janesha C Maddumage
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Sciences, La Trobe University, Victoria, 3086, Australia
| | - Eva Hesping
- Infectious Diseases & Immune Defense Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Sabrina Caiazzo
- Infectious Diseases & Immune Defense Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Keng Heng Lai
- Research Centre for Infectious Diseases, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Lee Ming Yeoh
- Burnet Institute, Melbourne, Victoria, 3004, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Joseph Menassa
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Sciences, La Trobe University, Victoria, 3086, Australia
| | - D Herbert Opi
- Burnet Institute, Melbourne, Victoria, 3004, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, 3052, Australia
- Central Clinical School and Department of Microbiology, Monash University, Clayton, Victoria, 3800, Australia
| | - Callum Cairns
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Sciences, La Trobe University, Victoria, 3086, Australia
| | - Hamsa Puthalakath
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Sciences, La Trobe University, Victoria, 3086, Australia
| | - James G Beeson
- Burnet Institute, Melbourne, Victoria, 3004, Australia
- Central Clinical School and Department of Microbiology, Monash University, Clayton, Victoria, 3800, Australia
- Department of Infectious Diseases, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Marc Kvansakul
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Sciences, La Trobe University, Victoria, 3086, Australia
| | - Justin A Boddey
- Infectious Diseases & Immune Defense Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Danny W Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, 5005, Australia
- Burnet Institute, Melbourne, Victoria, 3004, Australia
- Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Robin F Anders
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Sciences, La Trobe University, Victoria, 3086, Australia
| | - Michael Foley
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Sciences, La Trobe University, Victoria, 3086, Australia.
- AdAlta, Science Drive, Bundoora, Victoria, 3083, Australia.
| |
Collapse
|
2
|
Oboh MA, Asmorom N, Falade C, Ojurongbe O, Thomas BN. High genetic and haplotype diversity in vaccine candidate Pfceltos but not Pfrh5 among malaria-infected children in Ibadan, Nigeria. PeerJ 2023; 11:e16519. [PMID: 38099304 PMCID: PMC10720411 DOI: 10.7717/peerj.16519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 11/03/2023] [Indexed: 12/17/2023] Open
Abstract
Malaria remains a global public health challenge. The disease has a great impact in sub-Saharan Africa among children under five years of age and pregnant women. Malaria control programs targeting the parasite and mosquitoes vectors with combinational therapy and insecticide-treated bednets are becoming obsolete due to the phenomenon of resistance, which is a challenge for reducing morbidity and mortality. Malaria vaccines would be effective alternative to the problem of parasite and insecticide resistance, but focal reports of polymorphisms in malaria candidate antigens have made it difficult to design an effective malaria vaccine. Therefore, studies geared towards elucidating the polymorphic pattern and how genes targeted for vaccine design evolve are imperative. We have carried out molecular and genetic analysis of two genes encoding vaccine candidates-the Plasmodium falciparum cell traversal ookinetes and sporozoites (Pfceltos) and P. falciparum reticulocyte binding protein 5 (Pfrh5) in parasite isolates from malaria-infected children in Ibadan, Nigeria to evaluate their genetic diversity, relatedness and pattern of molecular evolution. Pfceltos and Pfrh5 genes were amplified from P. falciparum positive samples. Amplified fragments were purified and sequenced using the chain termination method. Post-sequence edit of fragments and application of various population genetic analyses was done. We observed a higher number of segregating sites and haplotypes in the Pfceltos than in Pfrh5 gene, the former also presenting higher haplotype (0.942) and nucleotide diversity (θ = 0.01219 and π = 0.01148). In contrast, a lower haplotype (0.426) and nucleotide diversity (θ = 0.00125; π = 0.00095) was observed in the Pfrh5 gene. Neutrality tests do not show deviation from neutral expectations for Pfceltos, with the circulation of multiple low frequency haplotypes (Tajima's D = -0.21637; Fu and Li's D = -0.08164; Fu and Li's F = -0.14051). Strong linkage disequilibrium was observed between variable sites, in each of the genes studied. We postulate that the high diversity and circulation of multiple haplotypes has the potential of making a Pfceltos-subunit vaccine ineffective, while the low genetic diversity of Pfrh5 gene substantiates its evolutionary conservation and potential as a malaria vaccine candidate.
Collapse
Affiliation(s)
- Mary Aigbiremo Oboh
- Biomedical Sciences, Rochester Institute of Technology, Rochester, NY, United States of America
| | - Naemy Asmorom
- Biomedical Sciences, Rochester Institute of Technology, Rochester, NY, United States of America
| | - Catherine Falade
- Pharmacology and Therapeutics, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Olusola Ojurongbe
- Medical Microbiology and Parasitology, Ladoke Akintola University of Technology, Ogbomosho, Osun, Nigeria
- Centre for Emerging and Re-emerging Infectious Diseases, Ladoke Akintola University of Technology, Ogbomosho, Oyo, Nigeria
| | - Bolaji N. Thomas
- Biomedical Sciences, Rochester Institute of Technology, Rochester, NY, United States of America
| |
Collapse
|
3
|
Perrotti E, L'Episcopia M, Menegon M, Soares IS, Rosas-Aguirre A, Speybroeck N, LLanos-Cuentas A, Menard D, Ferreira MU, Severini C. Reduced polymorphism of Plasmodium vivax early transcribed membrane protein (PvETRAMP) 11.2. Parasit Vectors 2023; 16:238. [PMID: 37461081 DOI: 10.1186/s13071-023-05851-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/27/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND ETRAMP11.2 (PVX_003565) is a well-characterized protein with antigenic potential. It is considered to be a serological marker for diagnostic tools, and it has been suggested as a potential vaccine candidate. Despite its immunological relevance, the polymorphism of the P. vivax ETRAMP11.2 gene (pvetramp11.2) remains undefined. The genetic variability of an antigen may limit the effectiveness of its application as a serological surveillance tool and in vaccine development and, therefore, the aim of this study was to investigate the genetic diversity of pvetramp11.2 in parasite populations from Amazonian regions and worldwide. We also evaluated amino acid polymorphism on predicted B-cell epitopes. The low variability of the sequence encoding PvETRAMP11.2 protein suggests that it would be a suitable marker in prospective serodiagnostic assays for surveillance strategies or in vaccine design against P. vivax malaria. METHODS The pvetramp11.2 of P. vivax isolates collected from Brazil (n = 68) and Peru (n = 36) were sequenced and analyzed to assess nucleotide polymorphisms, allele distributions, population differentiation, genetic diversity and signature of selection. In addition, sequences (n = 104) of seven populations from different geographical regions were retrieved from the PlasmoDB database and included in the analysis to study the worldwide allele distribution. Potential linear B-cell epitopes and their polymorphisms were also explored. RESULTS The multiple alignments of 208 pvetramp11.2 sequences revealed a low polymorphism and a marked geographical variation in allele diversity. Seven polymorphic sites and 11 alleles were identified. All of the alleles were detected in isolates from the Latin American region and five alleles were detected in isolates from the Southeast Asia/Papua New Guinea (SEA/PNG) region. Three alleles were shared by all Latin American populations (H1, H6 and H7). The H1 allele (reference allele from Salvador-1 strain), which was absent in the SEA/PNG populations, was the most represented allele in populations from Brazil (54%) and was also detected at high frequencies in populations from all other Latin America countries (range: 13.0% to 33.3%). The H2 allele was the major allele in SEA/PNG populations, but was poorly represented in Latin America populations (only in Brazil: 7.3%). Plasmodium vivax populations from Latin America showed a marked inter-population genetic differentiation (fixation index [Fst]) in contrast to SEA/PNG populations. Codon bias measures (effective number of codons [ENC] and Codon bias index [CBI]) indicated preferential use of synonymous codons, suggesting selective pressure at the translation level. Only three amino acid substitutions, located in the C-terminus, were detected. Linear B-cell epitope mapping predicted two epitopes in the Sal-1 PvETRAMP11.2 protein, one of which was fully conserved in all of the parasite populations analyzed. CONCLUSIONS We provide an overview of the allele distribution and genetic differentiation of ETRAMP11.2 antigen in P. vivax populations from different endemic areas of the world. The reduced polymorphism and the high degree of protein conservation supports the application of PvETRAMP11.2 protein as a reliable antigen for application in serological assays or vaccine design. Our findings provide useful information that can be used to inform future study designs.
Collapse
Affiliation(s)
- Edvige Perrotti
- Department of Infectious Diseases, Istituto Superiore Di Sanità, Rome, Italy.
| | | | - Michela Menegon
- Department of Infectious Diseases, Istituto Superiore Di Sanità, Rome, Italy
| | - Irene S Soares
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Angel Rosas-Aguirre
- Research Institute of Health and Society (IRSS), Université Catholique de Louvain, Brussels, Belgium
- Instituto de Medicina Tropical "Alexander Von Humboldt", Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Niko Speybroeck
- Research Institute of Health and Society (IRSS), Université Catholique de Louvain, Brussels, Belgium
| | - Alejandro LLanos-Cuentas
- Instituto de Medicina Tropical "Alexander Von Humboldt", Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Didier Menard
- Laboratoire de Parasitologie Et Mycologie Médicale, Les Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- Institut de Parasitologie Et Pathologie Tropicale, Université de Strasbourg, Strasbourg, France
- Malaria Genetics and Resistance Unit-INSERM U1201, Institut Pasteur, Paris, France
| | - Marcelo Urbano Ferreira
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Carlo Severini
- Department of Infectious Diseases, Istituto Superiore Di Sanità, Rome, Italy.
| |
Collapse
|
4
|
Defining species-specific and conserved interactions of apical membrane protein 1 during erythrocyte invasion in malaria to inform multi-species vaccines. Cell Mol Life Sci 2023; 80:74. [PMID: 36847896 PMCID: PMC9969379 DOI: 10.1007/s00018-023-04712-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/17/2023] [Accepted: 01/30/2023] [Indexed: 03/01/2023]
Abstract
Plasmodium falciparum and P. vivax are the major causes of human malaria, and P. knowlesi is an important additional cause in SE Asia. Binding of apical membrane antigen 1 (AMA1) to rhoptry neck protein 2 (RON2) was thought to be essential for merozoite invasion of erythrocytes by Plasmodium spp. Our findings reveal that P. falciparum and P. vivax have diverged and show species-specific binding of AMA1 to RON2, determined by a β-hairpin loop in RON2 and specific residues in AMA1 Loop1E. In contrast, cross-species binding of AMA1 to RON2 is retained between P. vivax and P. knowlesi. Mutation of specific amino acids in AMA1 Loop1E in P. falciparum or P. vivax ablated RON2 binding without impacting erythrocyte invasion. This indicates that the AMA1-RON2-loop interaction is not essential for invasion and additional AMA1 interactions are involved. Mutations in AMA1 that disrupt RON2 binding also enable escape of invasion inhibitory antibodies. Therefore, vaccines and therapeutics will need to be broader than targeting only the AMA1-RON2 interaction. Antibodies targeting AMA1 domain 3 had greater invasion-inhibitory activity when RON2-loop binding was ablated, suggesting this domain is a promising additional target for vaccine development. Targeting multiple AMA1 interactions involved in invasion may enable vaccines that generate more potent inhibitory antibodies and address the capacity for immune evasion. Findings on specific residues for invasion function and species divergence and conservation can inform novel vaccines and therapeutics against malaria caused by three species, including the potential for cross-species vaccines.
Collapse
|
5
|
Oboh MA, Isaac C, Schroeter MN, Morenikeji OB, Amambua-Ngwa A, Meremikwu MM, Thomas BN. Population genetic analysis of Plasmodium falciparum cell-traversal protein for ookinetes and sporozoite among malaria patients from southern Nigeria. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2022; 105:105369. [PMID: 36182066 DOI: 10.1016/j.meegid.2022.105369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 09/12/2022] [Accepted: 09/20/2022] [Indexed: 11/27/2022]
Abstract
Plasmodium falciparum immune escape mechanisms affect antigens being prioritized for vaccine design. As a result of the multiple surface antigens the parasite exhibits at different life cycle stages, designing a vaccine that would efficiently boost the immune system in clearing infections has been challenging. The P. falciparum cell-traversal protein for ookinetes and sporozoite (Pfceltos) is instrumental for ookinete traversal of the mosquito midgut and sporozoites invasion of the human liver cells. Pfceltos elicits both humoral and cellular immune response but has been reported with multiple single nucleotide polymorphisms in global isolates. A cross-sectional survey, conducted in southern Nigeria, between January-March 2021 recruited 283 individuals. Of this, 166 demonstrated P. falciparum infections (86 from Cross River and 80 from Edo), 48 (55.8%) while only 36 (45%) were amplified for Pfceltos gene from both sites respectively. Fifty amplified samples were sequenced and analysed for their diversity, polymorphisms and population structure of the gene. The number of segregating sites in Edo State was higher (34) than that of Cross River State. Though nucleotide diversity was higher for Edo compared to Cross River State (θw = 0.02505; π = 0.03993 versus θw = 0.00930; π = 0.01033 respectively), the reverse was the case for haplotype diversity (0.757 versus 0.890 for Edo and Cross River respectively). Of the twelve haplotypes observed from both states, only two (KASLPVEK and NAFLSFEK) were shared, with haplotype prevalence higher in Edo (16% and 36%) than Cross River (8% and 4%). The Tajima's D test was positive for both states, with Fst value showing a strong genetic differentiation (Fst = 0.25599), indicating the occurrence of balancing selection favoring haplotype circulation at a low frequency. The shared haplotypes, low Hst and Fst values presents a challenge to predict the extent of gene flow. High LD values present a grim public health consequence should a Pfceltos-conjugated vaccine be considered for prophylaxis in Nigeria.
Collapse
Affiliation(s)
- Mary A Oboh
- Department of Biomedical Sciences, Rochester Institute of Technology, Rochester, NY, United States of America.
| | - Clement Isaac
- Department of Zoology, Faculty of Life Sciences, Ambrose Ali University, Ekpoma, Nigeria
| | - Marissa N Schroeter
- Department of Biomedical Sciences, Rochester Institute of Technology, Rochester, NY, United States of America
| | - Olanrewaju B Morenikeji
- Division of Biological and Health Sciences, University of Pittsburgh, Bradford, PA 16701, United States of America
| | - Alfred Amambua-Ngwa
- Medical Research Council Unit, The Gambia, at the London School of Hygiene and Tropical Medicine, Fajara, the Gambia
| | - Martin M Meremikwu
- University of Calabar Teaching Hospital, Calabar, Cross River State, Nigeria
| | - Bolaji N Thomas
- Department of Biomedical Sciences, Rochester Institute of Technology, Rochester, NY, United States of America.
| |
Collapse
|
6
|
Zhang Y, Li D, Lu S, Zheng B. Toxoplasmosis vaccines: what we have and where to go? NPJ Vaccines 2022; 7:131. [PMID: 36310233 PMCID: PMC9618413 DOI: 10.1038/s41541-022-00563-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
Despite recent major advances in developing effective vaccines against toxoplasmosis, finding new protective vaccination strategies remains a challenging and elusive goal as it is critical to prevent the disease. Over the past few years, various experimental approaches have shown that developing an effective vaccine against T. gondii is achievable. However, more remains unknown due to its complicated life cycle, difficulties in clinical translation, and lack of a standardized platform. This minireview summarizes the recent advances in the development of T. gondii vaccines and the main obstacles to developing a safe, effective and durable T. gondii vaccine. The successes and failures in developing and testing vaccine candidates for the T. gondii vaccine are also discussed, which may facilitate the future development of T. gondii vaccines.
Collapse
Affiliation(s)
- Yizhuo Zhang
- grid.506977.a0000 0004 1757 7957Institute of Parasitic Diseases, School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China ,grid.506977.a0000 0004 1757 7957Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Dan Li
- grid.506977.a0000 0004 1757 7957Institute of Parasitic Diseases, School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China ,grid.506977.a0000 0004 1757 7957Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Shaohong Lu
- grid.506977.a0000 0004 1757 7957Institute of Parasitic Diseases, School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China ,grid.506977.a0000 0004 1757 7957Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China ,grid.506977.a0000 0004 1757 7957Key Laboratory of Bio-tech Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Bin Zheng
- grid.506977.a0000 0004 1757 7957Institute of Parasitic Diseases, School of Basic Medicine and Forensics, Hangzhou Medical College, Hangzhou, China ,grid.506977.a0000 0004 1757 7957Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China ,grid.506977.a0000 0004 1757 7957Key Laboratory of Bio-tech Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
7
|
Xu Y, Zhou Z, Brooks B, Ferguson T, Obliosca J, Huang J, Kaneko I, Iwanaga S, Yuda M, Tsuji Y, Zhang H, Luo CC, Jiang X, Kong XP, Tsuji M, Tison CK. Layer-by-Layer Delivery of Multiple Antigens Using Trimethyl Chitosan Nanoparticles as a Malaria Vaccine Candidate. Front Immunol 2022; 13:900080. [PMID: 36059505 PMCID: PMC9428560 DOI: 10.3389/fimmu.2022.900080] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
Developing a safe and effective malaria vaccine is critical to reducing the spread and resurgence of this deadly disease, especially in children. In recent years, vaccine technology has seen expanded development of subunit protein, peptide, and nucleic acid vaccines. This is due to their inherent safety, the ability to tailor their immune response, simple storage requirements, easier production, and lower expense compared to using attenuated and inactivated organism-based approaches. However, these new vaccine technologies generally have low efficacy. Subunit vaccines, due to their weak immunogenicity, often necessitate advanced delivery vectors and/or the use of adjuvants. A new area of vaccine development involves design of synthetic micro- and nano-particles and adjuvants that can stimulate immune cells directly through their physical and chemical properties. Further, the unique and complex life cycle of the Plasmodium organism, with multiple stages and varying epitopes/antigens presented by the parasite, is another challenge for malaria vaccine development. Targeting multistage antigens simultaneously is therefore critical for an effective malaria vaccine. Here, we rationally design a layer-by-layer (LbL) antigen delivery platform (we called LbL NP) specifically engineered for malaria vaccines. A biocompatible modified chitosan nanoparticle (trimethyl chitosan, TMC) was synthesized and utilized for LbL loading and release of multiple malaria antigens from pre-erythrocytic and erythrocytic stages. LbL NP served as antigen/protein delivery vehicles and were demonstrated to induce the highest Plasmodium falciparum Circumsporozoite Protein (PfCSP) specific T-cell responses in mice studies as compared to multiple controls. From immunogenicity studies, it was concluded that two doses of intramuscular injection with a longer interval (4 weeks) than traditional malaria vaccine candidate dosing would be the vaccination potential for LbL NP vaccine candidates. Furthermore, in PfCSP/Py parasite challenge studies we demonstrated protective efficacy using LbL NP. These LbL NP provided a significant adjuvant effect since they may induce innate immune response that led to a potent adaptive immunity to mediate non-specific anti-malarial effect. Most importantly, the delivery of CSP full-length protein stimulated long-lasting protective immune responses even after the booster immunization 4 weeks later in mice.
Collapse
Affiliation(s)
- Yang Xu
- Luna Labs USA, Biotech Group, Charlottesville, VA, United States
- *Correspondence: Yang Xu,
| | - Ziyou Zhou
- Luna Labs USA, Biotech Group, Charlottesville, VA, United States
| | - Brad Brooks
- Luna Labs USA, Biotech Group, Charlottesville, VA, United States
| | - Tammy Ferguson
- Luna Labs USA, Biotech Group, Charlottesville, VA, United States
| | - Judy Obliosca
- Luna Labs USA, Biotech Group, Charlottesville, VA, United States
| | - Jing Huang
- HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center, New York, NY, United States
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - Izumi Kaneko
- Department of Medical Zoology, Mie University Graduate School of Medicine, Mie, Japan
| | - Shiroh Iwanaga
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Masao Yuda
- Department of Medical Zoology, Mie University Graduate School of Medicine, Mie, Japan
| | - Yukiko Tsuji
- HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center, New York, NY, United States
| | - Huitang Zhang
- Department of Biochemistry and Molecular Pharmacology, New York University (NYU) Grossman School of Medicine, New York, NY, United States
| | - Christina C. Luo
- Department of Biochemistry and Molecular Pharmacology, New York University (NYU) Grossman School of Medicine, New York, NY, United States
| | - Xunqing Jiang
- Department of Biochemistry and Molecular Pharmacology, New York University (NYU) Grossman School of Medicine, New York, NY, United States
| | - Xiang-Peng Kong
- Department of Biochemistry and Molecular Pharmacology, New York University (NYU) Grossman School of Medicine, New York, NY, United States
| | - Moriya Tsuji
- HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center, New York, NY, United States
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | | |
Collapse
|
8
|
Kurtovic L, Drew DR, Dent AE, Kazura JW, Beeson JG. Antibody Targets and Properties for Complement-Fixation Against the Circumsporozoite Protein in Malaria Immunity. Front Immunol 2021; 12:775659. [PMID: 34925347 PMCID: PMC8671933 DOI: 10.3389/fimmu.2021.775659] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/15/2021] [Indexed: 01/02/2023] Open
Abstract
The Plasmodium falciparum circumsporozoite protein (CSP) forms the basis of leading subunit malaria vaccine candidates. However, the mechanisms and specific targets of immunity are poorly defined. Recent findings suggest that antibody-mediated complement-fixation and activation play an important role in immunity. Here, we investigated the regions of CSP targeted by functional complement-fixing antibodies and the antibody properties associated with this activity. We quantified IgG, IgM, and functional complement-fixing antibody responses to different regions of CSP among Kenyan adults naturally exposed to malaria (n=102) and using a series of rabbit vaccination studies. Individuals who acquired functional complement-fixing antibodies had higher IgG, IgM and IgG1 and IgG3 to CSP. Acquired complement-fixing antibodies targeted the N-terminal, central-repeat, and C-terminal regions of CSP, and positive responders had greater antibody breadth compared to those who were negative for complement-fixing antibodies (p<0.05). Using rabbit vaccinations as a model, we confirmed that IgG specific to the central-repeat and non-repeat regions of CSP could effectively fix complement. However, vaccination with near full length CSP in rabbits poorly induced antibodies to the N-terminal region compared to naturally-acquired immunity in humans. Poor induction of N-terminal antibodies was also observed in a vaccination study performed in mice. IgG and IgM to all three regions of CSP play a role in mediating complement-fixation, which has important implications for malaria vaccine development.
Collapse
Affiliation(s)
- Liriye Kurtovic
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - Damien R. Drew
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
| | - Arlene E. Dent
- Center for Global Health and Diseases, Case Western University, Cleveland, OH, United States
| | - James W. Kazura
- Center for Global Health and Diseases, Case Western University, Cleveland, OH, United States
| | - James G. Beeson
- Life Sciences, Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
- Department of Microbiology, Monash University, Clayton, VIC, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
9
|
Genetic Polymorphism and Natural Selection of Apical Membrane Antigen-1 in Plasmodium falciparum Isolates from Vietnam. Genes (Basel) 2021; 12:genes12121903. [PMID: 34946853 PMCID: PMC8701107 DOI: 10.3390/genes12121903] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/22/2021] [Accepted: 11/26/2021] [Indexed: 11/17/2022] Open
Abstract
Apical membrane antigen-1 of Plasmodium falciparum (PfAMA-1) is a leading malaria vaccine candidate antigen. However, the genetic diversity of pfama-1 and associated antigenic variation in global P. falciparum field isolates are major hurdles to the design of an efficacious vaccine formulated with this antigen. Here, we analyzed the genetic structure and the natural selection of pfama-1 in the P. falciparum population of Vietnam. A total of 37 distinct haplotypes were found in 131 P. falciparum Vietnamese isolates. Most amino acid changes detected in Vietnamese pfama-1 were localized in the ectodomain, domains I, II, and III. Overall patterns of major amino acid changes in Vietnamese pfama-1 were similar to those of global pfama-1, but the frequencies of the amino acid changes slightly differed by country. Novel amino acid changes were also identified in Vietnamese pfama-1. Vietnamese pfama-1 revealed relatively lower genetic diversity than currently analyzed pfama-1 in other geographical regions, and suggested a distinct genetic differentiation pattern. Evidence for natural selection was detected in Vietnamese pfama-1, but it showed purifying selection unlike the global pfama-1 analyzed so far. Recombination events were also found in Vietnamese pfama-1. Major amino acid changes that were commonly identified in global pfama-1 were mainly localized to predicted B-cell epitopes, RBC-binding sites, and IUR regions. These results provide important information for understanding the genetic nature of the Vietnamese pfama-1 population, and have significant implications for the design of a vaccine based on PfAMA-1.
Collapse
|
10
|
Opi DH, Kurtovic L, Chan JA, Horton JL, Feng G, Beeson JG. Multi-functional antibody profiling for malaria vaccine development and evaluation. Expert Rev Vaccines 2021; 20:1257-1272. [PMID: 34530671 DOI: 10.1080/14760584.2021.1981864] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION A vaccine would greatly accelerate current global efforts toward malaria elimination. While a partially efficacious vaccine has been achieved for Plasmodium falciparum, a major bottleneck in developing highly efficacious vaccines is a lack of reliable correlates of protection, and the limited application of assays that quantify functional immune responses to evaluate and down-select vaccine candidates in pre-clinical studies and clinical trials. AREAS COVERED In this review, we describe the important role of antibodies in immunity against malaria and detail the nature and functional activities of antibodies against the malaria-causing parasite. We highlight the growing understanding of antibody effector functions against malaria and in vitro assays to measure these functional antibody responses. We discuss the application of these assays to quantify antibody functions in vaccine development and evaluation. EXPERT OPINION It is becoming increasingly clear that multiple antibody effector functions are involved in immunity to malaria. Therefore, we propose that evaluating vaccine candidates needs to move beyond individual assays or measuring IgG magnitude alone. Instead, vaccine evaluation should incorporate the full breadth of antibody response types and harness a wider range of assays measuring functional antibody responses. We propose a 3-tier approach to implementing assays to inform vaccine evaluation.
Collapse
Affiliation(s)
- D Herbert Opi
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - Liriye Kurtovic
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia
| | - Jo-Anne Chan
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - Jessica L Horton
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - Gaoqian Feng
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia
| | - James G Beeson
- Life Sciences, Burnet Institute, Melbourne, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Australia.,Department of Medicine, The Doherty Institute, The University of Melbourne, Melbourne, Australia.,Department of Microbiology, Monash University, Clayton, Australia
| |
Collapse
|
11
|
Analysis of Genetic Diversity in Indian Isolates of Rhipicephalus microplus Based on Bm86 Gene Sequence. Vaccines (Basel) 2021; 9:vaccines9030194. [PMID: 33652549 PMCID: PMC7996562 DOI: 10.3390/vaccines9030194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/21/2021] [Accepted: 01/26/2021] [Indexed: 11/16/2022] Open
Abstract
The control of cattle tick, Rhipicephalus microplus, is focused on repeated use of acaricides. However, due to growing acaricide resistance and residues problem, immunization of animals along with limited use of effective acaricides is considered a suitable option for the control of tick infestations. To date, more than fifty vaccine candidates have been identified and tested worldwide, but two vaccines were developed using the extensively studied candidate, Bm86. The main reason for limited vaccine commercialization in other countries is genetic diversity in the Bm86 gene leading to considerable variation in vaccine efficacy. India, with 193.46 million cattle population distributed in 28 states and 9 union territories, is suffering from multiple tick infestation dominated by R. microplus. As R. microplus has developed multi-acaricide resistance, an efficacious vaccine may provide a sustainable intervention for tick control. Preliminary experiments revealed that the presently available commercial vaccine based on the BM86 gene is not efficacious against Indian strain. In concert with the principle of reverse vaccinology, genetic polymorphism of the Bm86 gene within Indian isolates of R. microplus was studied. A 578 bp conserved nucleotide sequences of Bm86 from 65 R. microplus isolates collected from 9 Indian states was sequenced and revealed 95.6-99.8% and 93.2-99.5% identity in nucleotides and amino acids sequences, respectively. The identities of nucleotides and deduced amino acids were 94.7-99.8% and 91.8-99.5%, respectively, between full-length sequence (orf) of the Bm86 gene of IVRI-I strain and published sequences of vaccine strains. Six nucleotides deletion were observed in Indian Bm86 sequences. Four B-cell epitopes (D519-K554, H563-Q587, C598-T606, T609-K623), which are present in the conserved region of the IVRI-I Bm86 sequence, were selected. The results confirm that the use of available commercial Bm86 vaccines is not a suitable option against Indian isolates of R. microplus. A country-specific multi-epitope Bm86 vaccine consisting of four specific B-cell epitopes along with candidate molecules, subolesin and tropomyosin in chimeric/co-immunization format may provide a sustainable option for implementation in an integrated tick management system.
Collapse
|
12
|
Milán-Noris EM, Monreal-Escalante E, Rosales-Mendoza S, Soria-Guerra RE, Radwan O, Juvik JA, Korban SS. An AMA1/MSP1 19 Adjuvanted Malaria Transplastomic Plant-Based Vaccine Induces Immune Responses in Test Animals. Mol Biotechnol 2020; 62:534-545. [PMID: 32870446 DOI: 10.1007/s12033-020-00271-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2020] [Indexed: 01/12/2023]
Abstract
Malaria is a tropical human disease, caused by protozoan parasites, wherein a significant number of the world's population is at risk. Annually, more than 219 million new cases are reported. Although there are prevention treatments, there are no highly and widely effective licensed anti-malarial vaccines available for use. Opportunities for utilization of plant-based vaccines as novel platforms for developing safe, reliable, and affordable treatments offer promise for developing such a vaccine against malaria. In this study, a Malchloroplast candidate vaccine was designed, composed of segments of AMA1 and MSP1 proteins, two epitopes of Plasmodium falciparum, along with a GK1 peptide from Taenia solium as adjuvant, and this was expressed in tobacco chloroplasts. Transplastomic tobacco lines were generated using biolistic transformation, and these were confirmed to carry the synthetic gene construct. Expression of the synthetic GK1 peptide was confirmed using RT-PCR and Western blots. Furthermore, the GK1 peptide was detected by HPLC at levels of up to 6 µg g-1 dry weight of tobacco leaf tissue. The plant-derived Malchloroplast candidate vaccine was subsequently tested in BALB/c female mice following subcutaneous administration, and was found to elicit specific humoral responses. Furthermore, components of this candidate vaccine were recognized by antibodies in Plasmodium falciparum malaria patients and were immunogenic in test mice. Thus, this study provided a 'proof of concept' for a promising plant-based candidate subunit vaccine against malaria.
Collapse
Affiliation(s)
- Evelia M Milán-Noris
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, Sinaloa, Mexico
| | - Elizabeth Monreal-Escalante
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, 78210, San Luis Potosi, SLP, Mexico
| | - Sergio Rosales-Mendoza
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, 78210, San Luis Potosi, SLP, Mexico.
| | - Ruth E Soria-Guerra
- Laboratorio de Ingeniería de Biorreactores, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, 78210, San Luis Potosi, SLP, Mexico
| | - Osman Radwan
- Department of Natural Resources and Environmental Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Environmental Microbiology Group, University of Dayton, Dayton, OH, 45469, USA
| | - John A Juvik
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Crop Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Schuyler S Korban
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Department of Natural Resources and Environmental Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
13
|
Wang YN, Lin M, Liang XY, Chen JT, Xie DD, Wang YL, Ehapo CS, Eyi UM, Huang HY, Wu JL, Xu DY, Chen ZM, Cao YL, Chen HB. Natural selection and genetic diversity of domain I of Plasmodium falciparum apical membrane antigen-1 on Bioko Island. Malar J 2019; 18:317. [PMID: 31533747 PMCID: PMC6751645 DOI: 10.1186/s12936-019-2948-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 09/06/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Plasmodium falciparum apical membrane antigen-1 (PfAMA-1) is a promising candidate antigen for a blood-stage malaria vaccine. However, antigenic variation and diversity of PfAMA-1 are still major problems to design a universal malaria vaccine based on this antigen, especially against domain I (DI). Detail understanding of the PfAMA-1 gene polymorphism can provide useful information on this potential vaccine component. Here, general characteristics of genetic structure and the effect of natural selection of DIs among Bioko P. falciparum isolates were analysed. METHODS 214 blood samples were collected from Bioko Island patients with P. falciparum malaria between 2011 and 2017. A fragment spanning DI of PfAMA-1 was amplified by nested polymerase chain reaction and sequenced. Polymorphic characteristics and the effect of natural selection were analysed using MEGA 5.0, DnaSP 6.0 and Popart programs. Genetic diversity in 576 global PfAMA-1 DIs were also analysed. Protein function prediction of new amino acid mutation sites was performed using PolyPhen-2 program. RESULTS 131 different haplotypes of PfAMA-1 were identified in 214 Bioko Island P. falciparum isolates. Most amino acid changes identified on Bioko Island were found in C1L. 32 amino acid changes identified in PfAMA-1 sequences from Bioko Island were found in predicted RBC-binding sites, B cell epitopes or IUR regions. Overall patterns of amino acid changes of Bioko PfAMA-1 DIs were similar to those in global PfAMA-1 isolates. Differential amino acid substitution frequencies were observed for samples from different geographical regions. Eight new amino acid changes of Bioko island isolates were also identified and their three-dimensional protein structural consequences were predicted. Evidence for natural selection and recombination event were observed in global isolates. CONCLUSIONS Patterns of nucleotide diversity and amino acid polymorphisms of Bioko Island isolates were similar to those of global PfAMA-1 DIs. Balancing natural selection across DIs might play a major role in generating genetic diversity in global isolates. Most amino acid changes in DIs occurred in predicted B-cell epitopes. Novel sites mapped on a three dimensional structure of PfAMA-1 showed that these regions were located at the corner. These results may provide significant value in the design of a malaria vaccine based on this antigen.
Collapse
Affiliation(s)
- Ya-Nan Wang
- Department of Histology and Embryology, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Min Lin
- School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou, Guangdong, People's Republic of China
| | - Xue-Yan Liang
- Department of Histology and Embryology, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
- School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou, Guangdong, People's Republic of China
| | - Jiang-Tao Chen
- Laboratory Medical Centre, Huizhou Municipal Central Hospital, Huizhou, Guangdong, People's Republic of China
- The Chinese Medical Aid Team to the Republic of Equatorial Guinea, Guangzhou, Guangdong, People's Republic of China
| | - Dong-De Xie
- Laboratory Medical Centre, Huizhou Municipal Central Hospital, Huizhou, Guangdong, People's Republic of China
| | - Yu-Ling Wang
- Laboratory Medical Centre, Huizhou Municipal Central Hospital, Huizhou, Guangdong, People's Republic of China
| | - Carlos Salas Ehapo
- Department of Medical Laboratory, Malabo Regional Hospital, Malabo, Equatorial Guinea
| | - Urbano Monsuy Eyi
- Department of Medical Laboratory, Malabo Regional Hospital, Malabo, Equatorial Guinea
| | - Hui-Ying Huang
- Department of Histology and Embryology, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
- School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou, Guangdong, People's Republic of China
| | - Jing-Li Wu
- 2014 Clinical Medicine Programme, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Dan-Yan Xu
- 2014 Clinical Medicine Programme, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Zhi-Mao Chen
- 2014 Clinical Medicine Programme, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Yi-Long Cao
- 2014 Clinical Medicine Programme, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Hai-Bin Chen
- Department of Histology and Embryology, Shantou University Medical College, Shantou, Guangdong, People's Republic of China.
| |
Collapse
|
14
|
Targets of complement-fixing antibodies in protective immunity against malaria in children. Nat Commun 2019; 10:610. [PMID: 30723225 PMCID: PMC6363798 DOI: 10.1038/s41467-019-08528-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/07/2019] [Indexed: 12/31/2022] Open
Abstract
Antibodies against P. falciparum merozoites fix complement to inhibit blood-stage replication in naturally-acquired and vaccine-induced immunity; however, specific targets of these functional antibodies and their importance in protective immunity are unknown. Among malaria-exposed individuals, we show that complement-fixing antibodies to merozoites are more strongly correlated with protective immunity than antibodies that inhibit growth quantified using the current reference assay for merozoite vaccine evaluation. We identify merozoite targets of complement-fixing antibodies and identify antigen-specific complement-fixing antibodies that are strongly associated with protection from malaria in a longitudinal study of children. Using statistical modelling, combining three different antigens targeted by complement-fixing antibodies could increase the potential protective effect to over 95%, and we identify antigens that were common in the most protective combinations. Our findings support antibody-complement interactions against merozoite antigens as important anti-malaria immune mechanisms, and identify specific merozoite antigens for further evaluation as vaccine candidates. Antibodies against Plasmodium falciparum merozoites that fix complement can inhibit blood-stage replication. Here, Reiling et al. show that complement-fixing antibodies strongly correlate with protective immunity in children, identify the merozoite targets, and predict antigen combinations that should result in strong protection.
Collapse
|
15
|
Pritam M, Singh G, Swaroop S, Singh AK, Singh SP. Exploitation of reverse vaccinology and immunoinformatics as promising platform for genome-wide screening of new effective vaccine candidates against Plasmodium falciparum. BMC Bioinformatics 2019; 19:468. [PMID: 30717656 PMCID: PMC7394322 DOI: 10.1186/s12859-018-2482-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 11/09/2018] [Indexed: 12/14/2022] Open
Abstract
Background In the current scenario, designing of world-wide effective malaria vaccine against Plasmodium falciparum remain challenging despite the significant progress has been made in last few decades. Conventional vaccinology (isolate, inactivate and inject) approaches are time consuming, laborious and expensive; therefore, the use of computational vaccinology tools are imperative, which can facilitate the design of new and promising vaccine candidates. Results In current investigation, initially 5548 proteins of P. falciparum genome were carefully chosen for the incidence of signal peptide/ anchor using SignalP4.0 tool that resulted into 640 surface linked proteins (SLP). Out of these SLP, only 17 were predicted to contain GPI-anchors using PredGPI tool in which further 5 proteins were considered as malarial antigenic adhesins by MAAP and VaxiJen programs, respectively. In the subsequent step, T cell epitopes of 5 genome derived predicted antigenic adhesins (GDPAA) and 5 randomly selected known malarial adhesins (RSKMA) were analysed employing MHC class I and II tools of IEDB analysis resource. Finally, VaxiJen scored T cell epitopes from each antigen were considered for prediction of population coverage (PPC) analysis in the world-wide population including malaria endemic regions. The validation of the present in silico strategy was carried out by comparing the PPC of combined (MHC class I and II) predicted epitope ensemble among GDPAA (99.97%), RSKMA (99.90%) and experimentally known epitopes (EKE) of P. falciparum (97.72%) pertaining to world-wide human population. Conclusions The present study systematically screened 5 potential protective antigens from P. falciparum genome using bioinformatics tools. Interestingly, these GDPAA, RSKMA and EKE of P. falciparum epitope ensembles forecasted to contain highly promiscuous T cell epitopes, which are potentially effective for most of the world-wide human population with malaria endemic regions. Therefore, these epitope ensembles could be considered in near future for novel and significantly effective vaccine candidate against malaria. Electronic supplementary material The online version of this article (10.1186/s12859-018-2482-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Manisha Pritam
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, 226028, India
| | - Garima Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, 226028, India
| | - Suchit Swaroop
- Department of Zoology, University of Lucknow, Lucknow, 226007, India
| | - Akhilesh Kumar Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, 226028, India
| | - Satarudra Prakash Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, 226028, India.
| |
Collapse
|
16
|
Beeson JG, Kurtovic L, Dobaño C, Opi DH, Chan JA, Feng G, Good MF, Reiling L, Boyle MJ. Challenges and strategies for developing efficacious and long-lasting malaria vaccines. Sci Transl Med 2019; 11:11/474/eaau1458. [DOI: 10.1126/scitranslmed.aau1458] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/05/2018] [Accepted: 11/02/2018] [Indexed: 12/24/2022]
Abstract
Although there has been major recent progress in malaria vaccine development, substantial challenges remain for achieving highly efficacious and durable vaccines against Plasmodium falciparum and Plasmodium vivax malaria. Greater knowledge of mechanisms and key targets of immunity are needed to accomplish this goal, together with new strategies for generating potent, long-lasting, functional immunity against multiple antigens. Implementation considerations in endemic areas will ultimately affect vaccine effectiveness, so innovations to simplify and enhance delivery are also needed. Whereas challenges remain, recent exciting progress and emerging knowledge promise hope for the future of malaria vaccines.
Collapse
|
17
|
Fuzzy cluster correlation mapping for online evaluation of teaching efficacy towards IoT study. COGN SYST RES 2018. [DOI: 10.1016/j.cogsys.2018.07.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
18
|
Afridi SG, Irfan M, Ahmad H, Aslam M, Nawaz M, Ilyas M, Khan A. Population genetic structure of domain I of apical membrane antigen-1 in Plasmodium falciparum isolates from Hazara division of Pakistan. Malar J 2018; 17:389. [PMID: 30367656 PMCID: PMC6203999 DOI: 10.1186/s12936-018-2539-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/20/2018] [Indexed: 11/10/2022] Open
Abstract
Background The Plasmodium falciparum apical membrane antigen-1 (PfAMA1) is considered as an ideal vaccine candidate for malaria control due to its high level of immunogenicity and essential role in parasite survival. Among the three domains of PfAMA1 protein, hyper-variable region (HVR) of domain I is the most immunogenic. The present study was conducted to evaluate the extent of genetic diversity across HVR domain I of the pfama1 gene in P. falciparum isolates from Hazara division of Pakistan. Methods The HVR domain I of the pfama1 was amplified and sequenced from 20 P. falciparum positive cases from Hazara division of Pakistan. The sequences were analysed in context of global population data of P. falciparum from nine malaria endemic countries. The DNA sequence reads quality assessment, reads assembling, sequences alignment/phylogenetic and population genetic analyses were performed using Staden, Lasergene v. 7.1, MEGA7 and DnaSP v.5 software packages respectively. Results Total 14 mutations were found in Pakistani isolates with 12 parsimony informative sites. During comparison with global isolates, a novel non-synonymous mutation (Y240F) was found specifically in a single Pakistani sample with 5% frequency. The less number of mutations, haplotypes, recombination and low pairwise nucleotide differences revealed tightly linked uniform genetic structure with low genetic diversity at HVR domain I of pfama1 among P. falciparum isolates from Hazara region of Pakistan. This uniform genetic structure may be shaped across Pakistani P. falciparum isolates by bottleneck or natural selection events. Conclusion The Pakistani P. falciparum isolates were found to maintain a distinct genetic pattern at HVR pfama1 with some extent of genetic relationship with geographically close Myanmar and Indian samples. However, the exact pattern of gene flow and demographic events may infer from whole genome sequence data with large sample size of P. falciparum collected from broad area of Pakistan. Electronic supplementary material The online version of this article (10.1186/s12936-018-2539-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sahib Gul Afridi
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Khyber Pakhtunkhwa, 23200, Pakistan
| | - Muhammad Irfan
- Center for Human Genetics, Hazara University, Mansehra, 21310, Pakistan
| | - Habib Ahmad
- Center for Human Genetics, Hazara University, Mansehra, 21310, Pakistan.,Center for OMIC Studies, Islamia College University, Peshawar, 25000, Pakistan
| | - Muneeba Aslam
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Khyber Pakhtunkhwa, 23200, Pakistan
| | - Mehwish Nawaz
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Khyber Pakhtunkhwa, 23200, Pakistan
| | - Muhammad Ilyas
- Center for Human Genetics, Hazara University, Mansehra, 21310, Pakistan
| | - Asifullah Khan
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Khyber Pakhtunkhwa, 23200, Pakistan.
| |
Collapse
|
19
|
Dominguez MF, González-Miguel J, Carmona C, Dalton JP, Cwiklinski K, Tort J, Siles-Lucas M. Low allelic diversity in vaccine candidates genes from different locations sustain hope for Fasciola hepatica immunization. Vet Parasitol 2018; 258:46-52. [PMID: 30105977 DOI: 10.1016/j.vetpar.2018.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/28/2018] [Accepted: 06/10/2018] [Indexed: 10/14/2022]
Abstract
Fasciola hepatica is a trematode parasite that causes fasciolosis in animals and humans. Fasciolosis is usually treated with triclabendazole, although drug-resistant parasites have been described in several geographical locations. An alternative to drug treatment would be the use of a vaccine, although vaccination studies that have been performed mainly in ruminants over the last 30 years, show high variability in the achieved protection and are not yet ready for commercialisation. Since F. hepatica exhibits a high degree of genomic polymorphism, variation in vaccine efficacy could be attributed, at least partially, to phenotypic differences in vaccine candidate sequences amongst parasites used in the challenge infections. To begin to address this issue, a collection of F. hepatica isolates from geographically dispersed regions, as well as parasites obtained from vaccination trials performed against a field isolate from Uruguay and the experimentally maintained South Gloucester isolate (Ridgeway Research, UK), were compiled to establish a F. hepatica Biobank. These collected isolates were used for the genetic analysis of several vaccine candidates that are important in host-parasite interactions and are the focus of the H2020 PARAGONE vaccine project (https://www.paragoneh2020.eu/), namely FhCL1, FhCL2, FhPrx, FhLAP and FhHDM. Our results show that F. hepatica exhibits a high level of conservation in the sequences encoding each of these proteins. The consequential low variability in these vaccine candidates amongst parasites from different geographical regions reinforces the idea that they would be suitable immunogens against liver fluke isolates worldwide.
Collapse
Affiliation(s)
- Maria Fernanda Dominguez
- Departamento de Genética, Facultad de Medicina, Universidad de la Republica, UDELAR, Montevideo, Uruguay
| | | | - Carlos Carmona
- Departamento de Genética, Facultad de Medicina, Universidad de la Republica, UDELAR, Montevideo, Uruguay
| | - John P Dalton
- School of Biological Sciences, Queen's University Belfast, United Kingdom
| | | | - José Tort
- Departamento de Genética, Facultad de Medicina, Universidad de la Republica, UDELAR, Montevideo, Uruguay
| | | |
Collapse
|
20
|
Feng G, Boyle MJ, Cross N, Chan JA, Reiling L, Osier F, Stanisic DI, Mueller I, Anders RF, McCarthy JS, Richards JS, Beeson JG. Human Immunization With a Polymorphic Malaria Vaccine Candidate Induced Antibodies to Conserved Epitopes That Promote Functional Antibodies to Multiple Parasite Strains. J Infect Dis 2018; 218:35-43. [PMID: 29584918 PMCID: PMC6904323 DOI: 10.1093/infdis/jiy170] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/22/2018] [Indexed: 12/15/2022] Open
Abstract
Background Overcoming antigenic diversity is a key challenge in the development of effective Plasmodium falciparum malaria vaccines. Strategies that promote the generation of antibodies targeting conserved epitopes of vaccine antigens may provide protection against diverse parasites strains. Understanding differences between vaccine-induced and naturally acquired immunity is important to achieving this goal. Methods We analyzed antibodies generated in a phase 1 human vaccine trial, MSP2-C1, which included 2 allelic forms of MSP2, an abundant vaccine antigen on the merozoite surface. Vaccine-induced responses were assessed for functional activity against multiple parasite strains, and cross-reactivity of antibodies was determined using competition ELISA and epitope mapping approaches. Results Vaccination induced cytophilic antibody responses with strain-transcending opsonic phagocytosis and complement-fixing function. In contrast to antibodies acquired via natural infection, vaccine-induced antibodies were directed towards conserved epitopes at the C-terminus of MSP2, whereas naturally acquired antibodies mainly targeted polymorphic epitopes. Functional activity of C-terminal-targeted antibodies was confirmed using monoclonal antibodies that promoted opsonic phagocytosis against multiple parasite strains. Conclusion Vaccination generated markedly different responses to polymorphic antigens than naturally acquired immunity and targeted conserved functional epitopes. Induction of antibodies targeting conserved regions of malaria antigens provides a promising vaccine strategy to overcome antigenic diversity for developing effective malaria vaccines.
Collapse
Affiliation(s)
- Gaoqian Feng
- Burnet Institute, Melbourne
- Department of Medicine, University of Melbourne, Parkville, Australia
| | | | | | | | | | - Faith Osier
- Burnet Institute, Melbourne
- Centre for Geographic Medicine - Coast, Kenya Medical Research Institute, Kilifi, Kenya
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Germany
| | | | - Ivo Mueller
- Walter and Eliza Hall Institute of Medical Research, Parkville
| | - Robin F Anders
- Department of Biochemistry and Genetics, La Trobe University, Melbourne
| | - James S McCarthy
- Clinical Tropical Medicine Laboratory, Queensland Institute of Medical Research Berghofer Medical Research Institute, Herston
| | - Jack S Richards
- Burnet Institute, Melbourne
- Department of Medicine, University of Melbourne, Parkville, Australia
- Central Clinical School and Department of Microbiology, Monash University, Melbourne, Australia
| | - James G Beeson
- Burnet Institute, Melbourne
- Department of Medicine, University of Melbourne, Parkville, Australia
- Central Clinical School and Department of Microbiology, Monash University, Melbourne, Australia
| |
Collapse
|
21
|
Early AM, Lievens M, MacInnis BL, Ockenhouse CF, Volkman SK, Adjei S, Agbenyega T, Ansong D, Gondi S, Greenwood B, Hamel M, Odero C, Otieno K, Otieno W, Owusu-Agyei S, Asante KP, Sorgho H, Tina L, Tinto H, Valea I, Wirth DF, Neafsey DE. Host-mediated selection impacts the diversity of Plasmodium falciparum antigens within infections. Nat Commun 2018; 9:1381. [PMID: 29643376 PMCID: PMC5895824 DOI: 10.1038/s41467-018-03807-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 03/14/2018] [Indexed: 12/28/2022] Open
Abstract
Host immunity exerts strong selective pressure on pathogens. Population-level genetic analysis can identify signatures of this selection, but these signatures reflect the net selective effect of all hosts and vectors in a population. In contrast, analysis of pathogen diversity within hosts provides information on individual, host-specific selection pressures. Here, we combine these complementary approaches in an analysis of the malaria parasite Plasmodium falciparum using haplotype sequences from thousands of natural infections in sub-Saharan Africa. We find that parasite genotypes show preferential clustering within multi-strain infections in young children, and identify individual amino acid positions that may contribute to strain-specific immunity. Our results demonstrate that natural host defenses to P. falciparum act in an allele-specific manner to block specific parasite haplotypes from establishing blood-stage infections. This selection partially explains the extreme amino acid diversity of many parasite antigens and suggests that vaccines targeting such proteins should account for allele-specific immunity. Host immune responses exert selective pressure on Plasmodium falciparum. Here, the authors show that allele-specific immunity impacts the antigenic diversity of individual malaria infections. This process partially explains the extreme amino acid diversity of many parasite antigens and suggests that vaccines should account for allele-specific immunity.
Collapse
Affiliation(s)
- Angela M Early
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA. .,Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.
| | | | - Bronwyn L MacInnis
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.,Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | | | - Sarah K Volkman
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.,Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.,Simmons College, School of Nursing and Health Sciences, Boston, MA, 02115, USA
| | - Samuel Adjei
- School of Medical Sciences, Kwame Nkrumah University of Science and Technology, KNUST - Kumasi, Ghana
| | - Tsiri Agbenyega
- School of Medical Sciences, Kwame Nkrumah University of Science and Technology, KNUST - Kumasi, Ghana
| | - Daniel Ansong
- School of Medical Sciences, Kwame Nkrumah University of Science and Technology, KNUST - Kumasi, Ghana
| | - Stacey Gondi
- KEMRI-Walter Reed Project, Kombewa, 40102, Kenya
| | - Brian Greenwood
- London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Mary Hamel
- KEMRI/CDC Research and Public Health Collaboration, Kisumu, 40100, Kenya
| | - Chris Odero
- KEMRI/CDC Research and Public Health Collaboration, Kisumu, 40100, Kenya
| | - Kephas Otieno
- KEMRI/CDC Research and Public Health Collaboration, Kisumu, 40100, Kenya
| | | | - Seth Owusu-Agyei
- London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK.,Kintampo Health Research Centre, Kintampo, 200, Ghana.,University of Health and Allied Science, PMB 31, Ho, Volta Region, Ghana
| | | | - Hermann Sorgho
- Institut de Recherche en Sciences de la Santé, Nanoro, Burkina Faso/Institute of Tropical Medicine, 2000, Antwerp, Belgium
| | - Lucas Tina
- KEMRI-Walter Reed Project, Kombewa, 40102, Kenya
| | - Halidou Tinto
- Institut de Recherche en Sciences de la Santé, Nanoro, Burkina Faso/Institute of Tropical Medicine, 2000, Antwerp, Belgium
| | - Innocent Valea
- Institut de Recherche en Sciences de la Santé, Nanoro, Burkina Faso/Institute of Tropical Medicine, 2000, Antwerp, Belgium
| | - Dyann F Wirth
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.,Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Daniel E Neafsey
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA. .,Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.
| |
Collapse
|
22
|
Abstract
Humoral immune responses against the malaria parasite are an important component of a protective immune response. Antibodies are often directed towards conformational epitopes, and the native structure of the antigenic region is usually critical for antibody recognition. We examined the structural features of various Plasmodium antigens that may impact on epitope location, by performing a comprehensive analysis of known and modelled structures from P. falciparum. Examining the location of known polymorphisms over all available structures, we observed a strong propensity for polymorphic residues to be exposed on the surface and to occur in particular secondary structure segments such as hydrogen-bonded turns. We also utilised established prediction algorithms for B-cell epitopes and MHC class II binding peptides, examining predicted epitopes in relation to known polymorphic sites within structured regions. Finally, we used the available structures to examine polymorphic hotspots and Tajima's D values using a spatial averaging approach. We identified a region of PfAMA1 involving both domains II and III under a high degree of balancing selection relative to the rest of the protein. In summary, we developed general methods for examining how sequence-based features relate to one another in three-dimensional space and applied these methods to key P. falciparum antigens.
Collapse
|
23
|
Kang JM, Lee J, Moe M, Jun H, Lê HG, Kim TI, Thái TL, Sohn WM, Myint MK, Lin K, Shin HJ, Kim TS, Na BK. Population genetic structure and natural selection of Plasmodium falciparum apical membrane antigen-1 in Myanmar isolates. Malar J 2018; 17:71. [PMID: 29415731 PMCID: PMC5804060 DOI: 10.1186/s12936-018-2215-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 01/30/2018] [Indexed: 12/16/2022] Open
Abstract
Background Plasmodium falciparum apical membrane antigen-1 (PfAMA-1) is one of leading blood stage malaria vaccine candidates. However, genetic variation and antigenic diversity identified in global PfAMA-1 are major hurdles in the development of an effective vaccine based on this antigen. In this study, genetic structure and the effect of natural selection of PfAMA-1 among Myanmar P. falciparum isolates were analysed. Methods Blood samples were collected from 58 Myanmar patients with falciparum malaria. Full-length PfAMA-1 gene was amplified by polymerase chain reaction and cloned into a TA cloning vector. PfAMA-1 sequence of each isolate was sequenced. Polymorphic characteristics and effect of natural selection were analysed with using DNASTAR, MEGA4, and DnaSP programs. Polymorphic nature and natural selection in 459 global PfAMA-1 were also analysed. Results Thirty-seven different haplotypes of PfAMA-1 were identified in 58 Myanmar P. falciparum isolates. Most amino acid changes identified in Myanmar PfAMA-1 were found in domains I and III. Overall patterns of amino acid changes in Myanmar PfAMA-1 were similar to those in global PfAMA-1. However, frequencies of amino acid changes differed by country. Novel amino acid changes in Myanmar PfAMA-1 were also identified. Evidences for natural selection and recombination event were observed in global PfAMA-1. Among 51 commonly identified amino acid changes in global PfAMA-1 sequences, 43 were found in predicted RBC-binding sites, B-cell epitopes, or IUR regions. Conclusions Myanmar PfAMA-1 showed similar patterns of nucleotide diversity and amino acid polymorphisms compared to those of global PfAMA-1. Balancing natural selection and intragenic recombination across PfAMA-1 are likely to play major roles in generating genetic diversity in global PfAMA-1. Most common amino acid changes in global PfAMA-1 were located in predicted B-cell epitopes where high levels of nucleotide diversity and balancing natural selection were found. These results highlight the strong selective pressure of host immunity on the PfAMA-1 gene. These results have significant implications in understanding the nature of Myanmar PfAMA-1 along with global PfAMA-1. They also provide useful information for the development of effective malaria vaccine based on this antigen.
Collapse
Affiliation(s)
- Jung-Mi Kang
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea.,BK21Plus Team for Anti-aging Biotechnology and Industry, Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Jinyoung Lee
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea.,Department of Infection Biology, Zoonosis Research Center, School of Medicine, Wonkwang University, Iksan, 54538, Republic of Korea
| | - Mya Moe
- Department of Medical Research Pyin Oo Lwin Branch, Pyin Oo Lwin, Myanmar
| | - Hojong Jun
- Department of Tropical Medicine and Inha Research Institute for Medical Sciences, Inha University College of Medicine, Incheon, 22212, Republic of Korea
| | - Hương Giang Lê
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea.,BK21Plus Team for Anti-aging Biotechnology and Industry, Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Tae Im Kim
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea.,Planning and Management Division, Nakdonggang National Institute of Biological Resources, Sangju, 37242, Republic of Korea
| | - Thị Lam Thái
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea.,BK21Plus Team for Anti-aging Biotechnology and Industry, Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Woon-Mok Sohn
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea
| | - Moe Kyaw Myint
- Department of Medical Research Pyin Oo Lwin Branch, Pyin Oo Lwin, Myanmar
| | - Khin Lin
- Department of Medical Research Pyin Oo Lwin Branch, Pyin Oo Lwin, Myanmar
| | - Ho-Joon Shin
- Department of Microbiology, Ajou University College of Medicine, Suwon, 16499, Republic of Korea
| | - Tong-Soo Kim
- Department of Tropical Medicine and Inha Research Institute for Medical Sciences, Inha University College of Medicine, Incheon, 22212, Republic of Korea
| | - Byoung-Kuk Na
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, 52727, Republic of Korea. .,BK21Plus Team for Anti-aging Biotechnology and Industry, Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea.
| |
Collapse
|
24
|
Miller RH, Hathaway NJ, Kharabora O, Mwandagalirwa K, Tshefu A, Meshnick SR, Taylor SM, Juliano JJ, Stewart VA, Bailey JA. A deep sequencing approach to estimate Plasmodium falciparum complexity of infection (COI) and explore apical membrane antigen 1 diversity. Malar J 2017; 16:490. [PMID: 29246158 PMCID: PMC5732508 DOI: 10.1186/s12936-017-2137-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 12/06/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Humans living in regions with high falciparum malaria transmission intensity harbour multi-strain infections comprised of several genetically distinct malaria haplotypes. The number of distinct malaria parasite haplotypes identified from an infected human host at a given time is referred to as the complexity of infection (COI). In this study, an amplicon-based deep sequencing method targeting the Plasmodium falciparum apical membrane antigen 1 (pfama1) was utilized to (1) investigate the relationship between P. falciparum prevalence and COI, (2) to explore the population genetic structure of P. falciparum parasites from malaria asymptomatic individuals participating in the 2007 Demographic and Health Survey (DHS) in the Democratic Republic of Congo (DRC), and (3) to explore selection pressures on geospatially divergent parasite populations by comparing AMA1 amino acid frequencies in the DRC and Mali. RESULTS A total of 900 P. falciparum infections across 11 DRC provinces were examined. Deep sequencing of both individuals, for COI analysis, and pools of individuals, to examine population structure, identified 77 unique pfama1 haplotypes. The majority of individual infections (64.5%) contained polyclonal (COI > 1) malaria infections based on the presence of genetically distinct pfama1 haplotypes. A minimal correlation between COI and malaria prevalence as determined by sensitive real-time PCR was identified. Population genetic analyses revealed extensive haplotype diversity, the vast majority of which was shared across the sites. AMA1 amino acid frequencies were similar between parasite populations in the DRC and Mali. CONCLUSIONS Amplicon-based deep sequencing is a useful tool for the detection of multi-strain infections that can aid in the understanding of antigen heterogeneity of potential malaria vaccine candidates, population genetics of malaria parasites, and factors that influence complex, polyclonal malaria infections. While AMA1 and other diverse markers under balancing selection may perform well for understanding COI, they may offer little geographic or temporal discrimination between parasite populations.
Collapse
Affiliation(s)
- Robin H Miller
- Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, USA
| | - Nicholas J Hathaway
- Program in Bioinformatics and Integrative Biology, University of Massachusetts School of Medicine, 55 Lake Avenue North, Worcester, MA, USA
| | - Oksana Kharabora
- University of North Carolina School of Medicine, 101 Manning Drive, Chapel Hill, NC, USA
| | - Kashamuka Mwandagalirwa
- Ecole de Santé Publique, Université de Kinshasa, Commune de Lemba, P.O Box 11850, Kinshasa, Democratic Republic of Congo
| | - Antoinette Tshefu
- Ecole de Santé Publique, Université de Kinshasa, Commune de Lemba, P.O Box 11850, Kinshasa, Democratic Republic of Congo
| | - Steven R Meshnick
- University of North Carolina School of Medicine, 101 Manning Drive, Chapel Hill, NC, USA
| | - Steve M Taylor
- Division of Infectious Diseases and Duke Global Health Institute, Duke University Medical Center, 303 Research Drive, Durham, NC, USA
| | - Jonathan J Juliano
- University of North Carolina School of Medicine, 101 Manning Drive, Chapel Hill, NC, USA
| | - V Ann Stewart
- Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, USA
| | - Jeffrey A Bailey
- Program in Bioinformatics and Integrative Biology, University of Massachusetts School of Medicine, 55 Lake Avenue North, Worcester, MA, USA.
| |
Collapse
|
25
|
Chan JA, Stanisic DI, Duffy MF, Robinson LJ, Lin E, Kazura JW, King CL, Siba PM, Fowkes FJ, Mueller I, Beeson JG. Patterns of protective associations differ for antibodies to P. falciparum-infected erythrocytes and merozoites in immunity against malaria in children. Eur J Immunol 2017; 47:2124-2136. [PMID: 28833064 DOI: 10.1002/eji.201747032] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 07/09/2017] [Accepted: 08/16/2017] [Indexed: 11/10/2022]
Abstract
Acquired antibodies play an important role in immunity to P. falciparum malaria and are typically directed towards surface antigens expressed by merozoites and infected erythrocytes (IEs). The importance of specific IE surface antigens as immune targets remains unclear. We evaluated antibodies and protective associations in two cohorts of children in Papua New Guinea. We used genetically-modified P. falciparum to evaluate the importance of PfEMP1 and a P. falciparum isolate with a virulent phenotype. Our findings suggested that PfEMP1 was the dominant target of antibodies to the IE surface, including functional antibodies that promoted opsonic phagocytosis by monocytes. Antibodies were associated with increasing age and concurrent parasitemia, and were higher among children exposed to a higher force-of-infection as determined using molecular detection. Antibodies to IE surface antigens were consistently associated with reduced risk of malaria in both younger and older children. However, protective associations for antibodies to merozoite surface antigens were only observed in older children. This suggests that antibodies to IE surface antigens, particularly PfEMP1, play an earlier role in acquired immunity to malaria, whereas greater exposure is required for protective antibodies to merozoite antigens. These findings have implications for vaccine design and serosurveillance of malaria transmission and immunity.
Collapse
Affiliation(s)
- Jo-Anne Chan
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia.,Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Danielle I Stanisic
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Institute for Glycomics, Griffith University, Southport, Queensland, Australia
| | - Michael F Duffy
- Department of Medicine and Melbourne School of Public Health, University of Melbourne, Parkville, Victoria, Australia
| | - Leanne J Robinson
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia.,Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Enmoore Lin
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - James W Kazura
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, USA
| | - Christopher L King
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, USA
| | - Peter M Siba
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Freya Ji Fowkes
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia.,Melbourne School of Public Health, University of Melbourne, Parkville, Victoria, Australia.,Department of Epidemiology and Preventive Medicine and Department of Infectious Diseases, Monash University, Melbourne, Victoria, Australia
| | - Ivo Mueller
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - James G Beeson
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia.,Department of Medicine and Melbourne School of Public Health, University of Melbourne, Parkville, Victoria, Australia.,Department of Microbiology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
26
|
Abstract
Evidence accumulated through the years clearly indicates that antiparasite immune responses can efficiently control malaria parasite infection at all development stages, and under certain circumstances they can prevent parasite infection. Translating these findings into vaccines or immunotherapeutic interventions has been difficult in part because of the extraordinary biological complexity of this parasite, which has several developmental stages expressing unique sets of stage-specific genes and multiple antigens, most of which are antigenically diverse. Nevertheless, in the last 30 years major advances have resulted in characterization of a number of vaccine candidates, exploration of the repertoire of host immune responses to the various parasite stages, and also identification of significant hurdles that need to be overcome. Most important, these advances strengthened the concept that the induction of host immune responses that target all developmental stages of Plasmodium can efficiently control or abrogate Plasmodium infections and strongly support the notion that an effective vaccine can be developed. This vaccine would be a critical component for programs aimed at controlling or eradicating malaria.
Collapse
Affiliation(s)
- Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institute of Health, Rockville, Maryland 20852
| | - Fidel Zavala
- Departmentof Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205
| |
Collapse
|
27
|
Srinivasan P, Baldeviano GC, Miura K, Diouf A, Ventocilla JA, Leiva KP, Lugo-Roman L, Lucas C, Orr-Gonzalez S, Zhu D, Villasante E, Soisson L, Narum DL, Pierce SK, Long CA, Diggs C, Duffy PE, Lescano AG, Miller LH. A malaria vaccine protects Aotus monkeys against virulent Plasmodium falciparum infection. NPJ Vaccines 2017; 2. [PMID: 28804644 PMCID: PMC5551459 DOI: 10.1038/s41541-017-0015-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The Plasmodium falciparum protein, apical membrane antigen 1 forms a complex with another parasite protein, rhoptry neck protein 2, to initiate junction formation with the erythrocyte and is essential for merozoite invasion during the blood stage of infection. Consequently, apical membrane antigen 1 has been a target of vaccine development but vaccination with apical membrane antigen 1 alone in controlled human malaria infections failed to protect and showed limited efficacy in field trials. Here we show that vaccination with AMA1–RON2L complex in Freund’s adjuvant protects Aotus monkeys against a virulent Plasmodium falciparum infection. Vaccination with AMA1 alone gave only partial protection, delaying infection in one of eight animals. However, the AMA1–RON2L complex vaccine completely protected four of eight monkeys and substantially delayed infection (>25 days) in three of the other four animals. Interestingly, antibodies from monkeys vaccinated with the AMA1–RON2L complex had significantly higher neutralizing activity than antibodies from monkeys vaccinated with AMA1 alone. Importantly, we show that antibodies from animals vaccinated with the complex have significantly higher neutralization activity against non-vaccine type parasites. We suggest that vaccination with the AMA1–RON2L complex induces functional antibodies that better recognize AMA1 as it appears complexed with RON2 during merozoite invasion. These data justify progression of this next generation AMA1 vaccine towards human trials. A vaccine targeting a protein complex that allows malaria-causing parasite to enter red blood cells has been produced. Malaria caused by the parasite Plasmodium falciparum is an oft-deadly infectious disease without an effective vaccine. A team of researchers at the National Institutes of Health led by Prakash Srinivasan, currently at the Johns Hopkins Malaria Research Institute, United States, demonstrated the efficacy of a vaccine candidate that works by priming a host’s immune system to a parasitic protein complex required to form a junction with red blood cells, allowing entry and proliferation of the pathogen. The group’s vaccine conferred more effective protection in monkeys than prior candidates that targeted only one component of the parasitic protein complex. This research warrants a closer look into how this candidate, and others targeting the protein complex, can be used to prevent malaria in humans.
Collapse
Affiliation(s)
- Prakash Srinivasan
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | | | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | | | - Karina P Leiva
- US Naval Medical Research Unit No. 6 (NAMRU-6), Callao, Peru
| | - Luis Lugo-Roman
- US Naval Medical Research Unit No. 6 (NAMRU-6), Callao, Peru
| | - Carmen Lucas
- US Naval Medical Research Unit No. 6 (NAMRU-6), Callao, Peru
| | - Sachy Orr-Gonzalez
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Daming Zhu
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Eileen Villasante
- US Military Malaria Vaccine Program, Naval Medical Research Center, Silver Spring, MD, USA
| | - Lorraine Soisson
- Malaria Vaccine Development Program, U.S. Agency for International Development, Washington, DC, USA
| | - David L Narum
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Susan K Pierce
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Carter Diggs
- Malaria Vaccine Development Program, U.S. Agency for International Development, Washington, DC, USA
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | | | - Louis H Miller
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
28
|
McCallum FJ, Persson KEM, Fowkes FJI, Reiling L, Mugyenyi CK, Richards JS, Simpson JA, Williams TN, Gilson PR, Hodder AN, Sanders PR, Anders RF, Narum DL, Chitnis C, Crabb BS, Marsh K, Beeson JG. Differing rates of antibody acquisition to merozoite antigens in malaria: implications for immunity and surveillance. J Leukoc Biol 2017; 101:913-925. [PMID: 27837017 PMCID: PMC5346181 DOI: 10.1189/jlb.5ma0716-294r] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 09/28/2016] [Accepted: 10/19/2016] [Indexed: 12/18/2022] Open
Abstract
Antibodies play a key role in acquired human immunity to Plasmodium falciparum (Pf) malaria and target merozoites to reduce or prevent blood-stage replication and the development of disease. Merozoites present a complex array of antigens to the immune system, and currently, there is only a partial understanding of the targets of protective antibodies and how responses to different antigens are acquired and boosted. We hypothesized that there would be differences in the rate of acquisition of antibodies to different antigens and how well they are boosted by infection, which impacts the acquisition of immunity. We examined responses to a range of merozoite antigens in 2 different cohorts of children and adults with different age structures and levels of malaria exposure. Overall, antibodies were associated with age, exposure, and active infection, and the repertoire of responses increased with age and active infection. However, rates of antibody acquisition varied between antigens and different regions within an antigen following exposure to malaria, supporting our hypothesis. Antigen-specific responses could be broadly classified into early response types in which antibodies were acquired early in childhood exposure and late response types that appear to require substantially more exposure for the development of substantial levels. We identified antigen-specific responses that were effectively boosted after recent infection, whereas other responses were not. These findings advance our understanding of the acquisition of human immunity to malaria and are relevant to the development of malaria vaccines targeting merozoite antigens and the selection of antigens for use in malaria surveillance.
Collapse
Affiliation(s)
- Fiona J McCallum
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
- Department of Drug Evaluation, Australian Army Malaria Institute, Brisbane, Australia
| | - Kristina E M Persson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Freya J I Fowkes
- Burnet Institute for Medical Research and Public Health, Melbourne, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Victoria, Australia
- Departments of Epidemiology and Preventive Medicine and Infectious Diseases, Monash University, Melbourne, Australia
| | - Linda Reiling
- Burnet Institute for Medical Research and Public Health, Melbourne, Australia
| | - Cleopatra K Mugyenyi
- Burnet Institute for Medical Research and Public Health, Melbourne, Australia
- Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Jack S Richards
- Burnet Institute for Medical Research and Public Health, Melbourne, Australia
- Department of Microbiology, Monash University, Melbourne, Australia
- Department of Medicine, University of Melbourne, Parkville, Australia
| | - Julie A Simpson
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Victoria, Australia
| | - Thomas N Williams
- Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Medicine, Imperial College of Science, Technology and Medicine, London, United Kingdom
| | - Paul R Gilson
- Burnet Institute for Medical Research and Public Health, Melbourne, Australia
| | - Anthony N Hodder
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Paul R Sanders
- Burnet Institute for Medical Research and Public Health, Melbourne, Australia
| | - Robin F Anders
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, Latrobe University, Melbourne, Australia
| | - David L Narum
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Brendan S Crabb
- Burnet Institute for Medical Research and Public Health, Melbourne, Australia
| | - Kevin Marsh
- Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
- Nuffield Department of Medicine, Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Oxford, United Kingdom
| | - James G Beeson
- Burnet Institute for Medical Research and Public Health, Melbourne, Australia;
- Department of Microbiology, Monash University, Melbourne, Australia
- Department of Medicine, University of Melbourne, Parkville, Australia
| |
Collapse
|
29
|
Tham WH, Beeson JG, Rayner JC. Plasmodium vivax vaccine research - we've only just begun. Int J Parasitol 2016; 47:111-118. [PMID: 27899329 DOI: 10.1016/j.ijpara.2016.09.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 09/24/2016] [Accepted: 09/29/2016] [Indexed: 10/25/2022]
Abstract
Plasmodium vivax parasites cause the majority of malaria cases outside Africa, and are increasingly being acknowledged as a cause of severe disease. The unique attributes of P. vivax biology, particularly the capacity of the dormant liver stage, the hypnozoite, to maintain blood-stage infections even in the absence of active transmission, make blood-stage vaccines particularly attractive for this species. However, P. vivax vaccine development remains resolutely in first gear, with only a single blood-stage candidate having been evaluated in any depth. Experience with Plasmodium falciparum suggests that a much broader search for new candidates and a deeper understanding of high priority targets will be required to make significant advances. This review discusses some of the particular challenges of P. vivax blood-stage vaccine development, highlighting both recent advances and key remaining barriers to overcome in order to move development forward.
Collapse
Affiliation(s)
- Wai-Hong Tham
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - James G Beeson
- Macfarlane Burnet Institute of Medical Research, 85 Commercial Road, Melbourne, Victoria 3004, Australia; Central Clinical School and Department of Microbiology, Monash University, Victoria, Australia
| | - Julian C Rayner
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, United Kingdom.
| |
Collapse
|
30
|
Chan JA, Howell KB, Langer C, Maier AG, Hasang W, Rogerson SJ, Petter M, Chesson J, Stanisic DI, Duffy MF, Cooke BM, Siba PM, Mueller I, Bull PC, Marsh K, Fowkes FJI, Beeson JG. A single point in protein trafficking by Plasmodium falciparum determines the expression of major antigens on the surface of infected erythrocytes targeted by human antibodies. Cell Mol Life Sci 2016; 73:4141-58. [PMID: 27193441 PMCID: PMC5042999 DOI: 10.1007/s00018-016-2267-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/22/2016] [Accepted: 05/06/2016] [Indexed: 11/30/2022]
Abstract
Antibodies to blood-stage antigens of Plasmodium falciparum play a pivotal role in human immunity to malaria. During parasite development, multiple proteins are trafficked from the intracellular parasite to the surface of P. falciparum-infected erythrocytes (IEs). However, the relative importance of different proteins as targets of acquired antibodies, and key pathways involved in trafficking major antigens remain to be clearly defined. We quantified antibodies to surface antigens among children, adults, and pregnant women from different malaria-exposed regions. We quantified the importance of antigens as antibody targets using genetically engineered P. falciparum with modified surface antigen expression. Genetic deletion of the trafficking protein skeleton-binding protein-1 (SBP1), which is involved in trafficking the surface antigen PfEMP1, led to a dramatic reduction in antibody recognition of IEs and the ability of human antibodies to promote opsonic phagocytosis of IEs, a key mechanism of parasite clearance. The great majority of antibody epitopes on the IE surface were SBP1-dependent. This was demonstrated using parasite isolates with different genetic or phenotypic backgrounds, and among antibodies from children, adults, and pregnant women in different populations. Comparisons of antibody reactivity to parasite isolates with SBP1 deletion or inhibited PfEMP1 expression suggest that PfEMP1 is the dominant target of acquired human antibodies, and that other P. falciparum IE surface proteins are minor targets. These results establish SBP1 as part of a critical pathway for the trafficking of major surface antigens targeted by human immunity, and have key implications for vaccine development, and quantifying immunity in populations.
Collapse
Affiliation(s)
- Jo-Anne Chan
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, VIC, 3001, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Katherine B Howell
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Christine Langer
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, VIC, 3001, Australia
| | - Alexander G Maier
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Wina Hasang
- Department of Medicine, University of Melbourne, Parkville, VIC, Australia
| | - Stephen J Rogerson
- Department of Medicine, University of Melbourne, Parkville, VIC, Australia
| | - Michaela Petter
- Department of Medicine, University of Melbourne, Parkville, VIC, Australia
| | - Joanne Chesson
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | | | - Michael F Duffy
- Department of Medicine, University of Melbourne, Parkville, VIC, Australia
| | - Brian M Cooke
- Programs in Infection and Immunity and Cardiovascular Disease, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Department of Microbiology, Monash University, Melbourne, VIC, Australia
| | - Peter M Siba
- Papua New Guinea Institute of Medical Research, Madang, Papua New Guinea
| | - Ivo Mueller
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Peter C Bull
- Centre for Geographic Medicine Research, Coast, Kenya Medical Research Institute, Kilifi, Kenya
| | - Kevin Marsh
- Centre for Geographic Medicine Research, Coast, Kenya Medical Research Institute, Kilifi, Kenya
| | - Freya J I Fowkes
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, VIC, 3001, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Melbourne School of Public Health, University of Melbourne, Parkville, VIC, Australia
- Department of Epidemiology and Preventive Medicine and Department of Infectious Diseases, Monash University, Melbourne, VIC, Australia
| | - James G Beeson
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, VIC, 3001, Australia.
- Department of Medicine, University of Melbourne, Parkville, VIC, Australia.
- Department of Microbiology, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
31
|
Sedegah M, Peters B, Hollingdale MR, Ganeshan HD, Huang J, Farooq F, Belmonte MN, Belmonte AD, Limbach KJ, Diggs C, Soisson L, Chuang I, Villasante ED. Vaccine Strain-Specificity of Protective HLA-Restricted Class 1 P. falciparum Epitopes. PLoS One 2016; 11:e0163026. [PMID: 27695088 PMCID: PMC5047630 DOI: 10.1371/journal.pone.0163026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 09/01/2016] [Indexed: 11/19/2022] Open
Abstract
A DNA prime/adenovirus boost malaria vaccine encoding Plasmodium falciparum strain 3D7 CSP and AMA1 elicited sterile clinical protection associated with CD8+ T cell interferon-gamma (IFN-γ) cells responses directed to HLA class 1-restricted AMA1 epitopes of the vaccine strain 3D7. Since a highly effective malaria vaccine must be broadly protective against multiple P. falciparum strains, we compared these AMA1 epitopes of two P. falciparum strains (7G8 and 3D7), which differ by single amino acid substitutions, in their ability to recall CD8+ T cell activities using ELISpot and flow cytometry/intracellular staining assays. The 7G8 variant peptides did not recall 3D7 vaccine-induced CD8+ T IFN-γ cell responses in these assays, suggesting that protection may be limited to the vaccine strain. The predicted MHC binding affinities of the 7G8 variant epitopes were similar to the 3D7 epitopes, suggesting that the amino acid substitutions of the 7G8 variants may have interfered with TCR recognition of the MHC:peptide complex or that the 7G8 variant may have acted as an altered peptide ligand. These results stress the importance of functional assays in defining protective epitopes. Clinical Trials Registrations: NCT00870987, NCT00392015
Collapse
Affiliation(s)
- Martha Sedegah
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, 20910, United States of America
| | - Bjoern Peters
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, 92037, United States of America
| | - Michael R. Hollingdale
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, 20910, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Rockville, MD, 20817, United States of America
- * E-mail:
| | - Harini D. Ganeshan
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, 20910, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Rockville, MD, 20817, United States of America
| | - Jun Huang
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, 20910, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Rockville, MD, 20817, United States of America
| | - Fouzia Farooq
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, 20910, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Rockville, MD, 20817, United States of America
| | - Maria N. Belmonte
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, 20910, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Rockville, MD, 20817, United States of America
| | - Arnel D. Belmonte
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, 20910, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Rockville, MD, 20817, United States of America
| | - Keith J. Limbach
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, 20910, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Rockville, MD, 20817, United States of America
| | - Carter Diggs
- USAID, Washington, DC, 20523, United States of America
| | | | - Ilin Chuang
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, 20910, United States of America
| | - Eileen D. Villasante
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, 20910, United States of America
| |
Collapse
|
32
|
Drew DR, Wilson DW, Elliott SR, Cross N, Terheggen U, Hodder AN, Siba PM, Chelimo K, Dent AE, Kazura JW, Mueller I, Beeson JG. A novel approach to identifying patterns of human invasion-inhibitory antibodies guides the design of malaria vaccines incorporating polymorphic antigens. BMC Med 2016; 14:144. [PMID: 27658419 PMCID: PMC5034621 DOI: 10.1186/s12916-016-0691-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 09/08/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The polymorphic nature of many malaria vaccine candidates presents major challenges to achieving highly efficacious vaccines. Presently, there is very little knowledge on the prevalence and patterns of functional immune responses to polymorphic vaccine candidates in populations to guide vaccine design. A leading polymorphic vaccine candidate against blood-stage Plasmodium falciparum is apical membrane antigen 1 (AMA1), which is essential for erythrocyte invasion. The importance of AMA1 as a target of acquired human inhibitory antibodies, their allele specificity and prevalence in populations is unknown, but crucial for vaccine design. METHODS P. falciparum lines expressing different AMA1 alleles were genetically engineered and used to quantify functional antibodies from two malaria-exposed populations of adults and children. The acquisition of AMA1 antibodies was also detected using enzyme-linked immunosorbent assay (ELISA) and competition ELISA (using different AMA1 alleles) from the same populations. RESULTS We found that AMA1 was a major target of naturally acquired invasion-inhibitory antibodies that were highly prevalent in malaria-endemic populations and showed a high degree of allele specificity. Significantly, the prevalence of inhibitory antibodies to different alleles varied substantially within populations and between geographic locations. Inhibitory antibodies to three specific alleles were highly prevalent (FVO and W2mef in Papua New Guinea; FVO and XIE in Kenya), identifying them for potential vaccine inclusion. Measurement of antibodies by standard or competition ELISA was not strongly predictive of allele-specific inhibitory antibodies. The patterns of allele-specific functional antibody responses detected with our novel assays may indicate that acquired immunity is elicited towards serotypes that are prevalent in each geographic location. CONCLUSIONS These findings provide new insights into the nature and acquisition of functional immunity to a polymorphic vaccine candidate and strategies to quantify functional immunity in populations to guide rational vaccine design.
Collapse
Affiliation(s)
- Damien R Drew
- The Burnet Institute of Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, 3004, Australia
| | - Danny W Wilson
- The Burnet Institute of Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, 3004, Australia.,Research Centre for Infectious Diseases, School of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
| | - Salenna R Elliott
- The Burnet Institute of Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, 3004, Australia
| | - Nadia Cross
- The Burnet Institute of Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, 3004, Australia
| | - Ulrich Terheggen
- The Burnet Institute of Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, 3004, Australia.,Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Anthony N Hodder
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Peter M Siba
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | | | - Arlene E Dent
- Case Western Reserve University, Cleveland, Ohio, USA
| | | | - Ivo Mueller
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - James G Beeson
- The Burnet Institute of Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, 3004, Australia. .,Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia. .,Department of Microbiology, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
33
|
Clark EL, Tomley FM, Blake DP. Are Eimeria Genetically Diverse, and Does It Matter? Trends Parasitol 2016; 33:231-241. [PMID: 27593338 DOI: 10.1016/j.pt.2016.08.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 08/05/2016] [Accepted: 08/09/2016] [Indexed: 10/21/2022]
Abstract
Eimeria pose a risk to all livestock species as a cause of coccidiosis, reducing productivity and compromising animal welfare. Pressure to reduce drug use in the food chain makes the development of cost-effective vaccines against Eimeria essential. For novel vaccines to be successful, understanding genetic and antigenic diversity in field populations is key. Eimeria species that infect chickens are most significant, with Eimeria tenella among the best studied and most economically important. Genome-wide single nucleotide polymorphism (SNP)-based haplotyping has been used to determine population structure, genotype distribution, and potential for cross-fertilization between E. tenella strains. Here, we discuss recent developments in our understanding of diversity for Eimeria in relation to its specialized life cycle, distribution across the globe, and the challenges posed to vaccine development.
Collapse
Affiliation(s)
- Emily L Clark
- Department of Pathology and Pathogen Biology, Royal Veterinary College, North Mymms, Hertfordshire, UK; Current address: The Roslin Institute, The University of Edinburgh, Easter Bush, Midlothian, UK
| | - Fiona M Tomley
- Department of Pathology and Pathogen Biology, Royal Veterinary College, North Mymms, Hertfordshire, UK
| | - Damer P Blake
- Department of Pathology and Pathogen Biology, Royal Veterinary College, North Mymms, Hertfordshire, UK.
| |
Collapse
|
34
|
Murungi LM, Sondén K, Llewellyn D, Rono J, Guleid F, Williams AR, Ogada E, Thairu A, Färnert A, Marsh K, Draper SJ, Osier FHA. Targets and Mechanisms Associated with Protection from Severe Plasmodium falciparum Malaria in Kenyan Children. Infect Immun 2016; 84:950-963. [PMID: 26787721 PMCID: PMC4807498 DOI: 10.1128/iai.01120-15] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 01/14/2016] [Indexed: 01/23/2023] Open
Abstract
Severe malaria (SM) is a life-threatening complication of infection with Plasmodium falciparum Epidemiological observations have long indicated that immunity against SM is acquired relatively rapidly, but prospective studies to investigate its immunological basis are logistically challenging and have rarely been undertaken. We investigated the merozoite targets and antibody-mediated mechanisms associated with protection against SM in Kenyan children aged 0 to 2 years. We designed a unique prospective matched case-control study of well-characterized SM clinical phenotypes nested within a longitudinal birth cohort of children (n= 5,949) monitored over the first 2 years of life. We quantified immunological parameters in sera collected before the SM event in cases and their individually matched controls to evaluate the prospective odds of developing SM in the first 2 years of life. Anti-AMA1 antibodies were associated with a significant reduction in the odds of developing SM (odds ratio [OR] = 0.37; 95% confidence interval [CI] = 0.15 to 0.90; P= 0.029) after adjustment for responses to all other merozoite antigens tested, while those against MSP-2, MSP-3, Plasmodium falciparum Rh2 [PfRh2], MSP-119, and the infected red blood cell surface antigens were not. The combined ability of total IgG to inhibit parasite growth and mediate the release of reactive oxygen species from neutrophils was associated with a marked reduction in the odds of developing SM (OR = 0.07; 95% CI = 0.006 to 0.82;P= 0.03). Assays of these two functional mechanisms were poorly correlated (Spearman rank correlation coefficient [rs] = 0.12;P= 0.07). Our data provide epidemiological evidence that multiple antibody-dependent mechanisms contribute to protective immunity via distinct targets whose identification could accelerate the development of vaccines to protect against SM.
Collapse
Affiliation(s)
- Linda M Murungi
- KEMRI Wellcome Trust Research Programme, Centre for Geographical Medicine Research-Coast, Kilifi, Kenya
| | - Klara Sondén
- Unit of Infectious Diseases, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - David Llewellyn
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Josea Rono
- KEMRI Wellcome Trust Research Programme, Centre for Geographical Medicine Research-Coast, Kilifi, Kenya
| | - Fatuma Guleid
- KEMRI Wellcome Trust Research Programme, Centre for Geographical Medicine Research-Coast, Kilifi, Kenya
| | | | - Edna Ogada
- KEMRI Wellcome Trust Research Programme, Centre for Geographical Medicine Research-Coast, Kilifi, Kenya
| | - Amos Thairu
- KEMRI Wellcome Trust Research Programme, Centre for Geographical Medicine Research-Coast, Kilifi, Kenya
| | - Anna Färnert
- Unit of Infectious Diseases, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Kevin Marsh
- KEMRI Wellcome Trust Research Programme, Centre for Geographical Medicine Research-Coast, Kilifi, Kenya
- Nuffield Department of Medicine, Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Churchill Hospital, Oxford, United Kingdom
| | - Simon J Draper
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Faith H A Osier
- KEMRI Wellcome Trust Research Programme, Centre for Geographical Medicine Research-Coast, Kilifi, Kenya
| |
Collapse
|
35
|
Weiss GE, Crabb BS, Gilson PR. Overlaying Molecular and Temporal Aspects of Malaria Parasite Invasion. Trends Parasitol 2016; 32:284-295. [DOI: 10.1016/j.pt.2015.12.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 12/02/2015] [Accepted: 12/10/2015] [Indexed: 12/31/2022]
|
36
|
Al-Qahtani AA, Abdel-Muhsin AMA, Dajem SMB, AlSheikh AAH, Bohol MFF, Al-Ahdal MN, Putaporntip C, Jongwutiwes S. Comparative sequence analysis of domain I of Plasmodium falciparum apical membrane antigen 1 from Saudi Arabia and worldwide isolates. INFECTION GENETICS AND EVOLUTION 2016; 39:381-388. [PMID: 26867816 DOI: 10.1016/j.meegid.2016.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 02/05/2016] [Accepted: 02/06/2016] [Indexed: 10/22/2022]
Abstract
The apical membrane antigen 1 of Plasmodium falciparum (PfAMA1) plays a crucial role in erythrocyte invasion and is a target of protective antibodies. Although domain I of PfAMA1 has been considered a promising vaccine component, extensive sequence diversity in this domain could compromise an effective vaccine design. To explore the extent of sequence diversity in domain I of PfAMA1, P. falciparum-infected blood samples from Saudi Arabia collected between 2007 and 2009 were analyzed and compared with those from worldwide parasite populations. Forty-six haplotypes and a novel codon change (M190V) were found among Saudi Arabian isolates. The haplotype diversity (0.948±0.004) and nucleotide diversity (0.0191±0.0008) were comparable to those from African hyperendemic countries. Positive selection in domain I of PfAMA1 among Saudi Arabian parasite population was observed because nonsynonymous nucleotide substitutions per nonsynonymous site (dN) significantly exceeded synonymous nucleotide substitutions per synonymous site (dS) and Tajima's D and its related statistics significantly deviated from neutrality in the positive direction. Despite a relatively low prevalence of malaria in Saudi Arabia, a minimum of 17 recombination events occurred in domain I. Genetic differentiation was significant between P. falciparum in Saudi Arabia and parasites from other geographic origins. Several shared or closely related haplotypes were found among parasites from different geographic areas, suggesting that vaccine derived from multiple shared epitopes could be effective across endemic countries.
Collapse
Affiliation(s)
- Ahmed A Al-Qahtani
- Department of Infection and Immunity, King Faisal Specialist Hospital & Research Center, Riyadh 11211, Saudi Arabia; Department of Microbiology and Immunology, Alfaisal University College of Medicine, Riyadh, Saudi Arabia.
| | - Abdel-Muhsin A Abdel-Muhsin
- Tropical Medicine Research Institute, National Centre for Research, Sudan; Department of Biology, Faculty of Science, University of Hail, Hail, Saudi Arabia
| | - Saad M Bin Dajem
- Department of Biology, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | - Adel Ali H AlSheikh
- National Center for Vector-Borne Diseases, Ministry of Health, Jazan, Saudi Arabia
| | - Marie Fe F Bohol
- Department of Infection and Immunity, King Faisal Specialist Hospital & Research Center, Riyadh 11211, Saudi Arabia
| | - Mohammed N Al-Ahdal
- Department of Infection and Immunity, King Faisal Specialist Hospital & Research Center, Riyadh 11211, Saudi Arabia; Department of Microbiology and Immunology, Alfaisal University College of Medicine, Riyadh, Saudi Arabia
| | - Chaturong Putaporntip
- Molecular Biology of Malaria and Opportunistic Parasites Research Unit, Department of Parasitology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Somchai Jongwutiwes
- Molecular Biology of Malaria and Opportunistic Parasites Research Unit, Department of Parasitology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW Malaria is caused by the infection and proliferation of parasites from the genus Plasmodium in red blood cells (RBCs). A free Plasmodium parasite, or merozoite, released from an infected RBC must invade another RBC host cell to sustain a blood-stage infection. Here, we review recent advances on RBC invasion by Plasmodium merozoites, focusing on specific molecular interactions between host and parasite. RECENT FINDINGS Recent work highlights the central role of host-parasite interactions at virtually every stage of RBC invasion by merozoites. Biophysical experiments have for the first time measured the strength of merozoite-RBC attachment during invasion. For P. falciparum, there have been many key insights regarding the invasion ligand PfRh5 in particular, including its influence on host species tropism, a co-crystal structure with its RBC receptor basigin, and its suitability as a vaccine target. For P. vivax, researchers identified the origin and emergence of the parasite from Africa, demonstrating a natural link to the Duffy-negative RBC variant in African populations. For the simian parasite P. knowlesi, zoonotic invasion into human cells is linked to RBC age, which has implications for parasitemia during an infection and thus malaria. SUMMARY New studies of the molecular and cellular mechanisms governing RBC invasion by Plasmodium parasites have shed light on various aspects of parasite biology and host cell tropism, and indicate opportunities for malaria control.
Collapse
|
38
|
Beeson JG, Drew DR, Boyle MJ, Feng G, Fowkes FJI, Richards JS. Merozoite surface proteins in red blood cell invasion, immunity and vaccines against malaria. FEMS Microbiol Rev 2016; 40:343-72. [PMID: 26833236 PMCID: PMC4852283 DOI: 10.1093/femsre/fuw001] [Citation(s) in RCA: 242] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2016] [Indexed: 01/11/2023] Open
Abstract
Malaria accounts for an enormous burden of disease globally, with Plasmodium falciparum accounting for the majority of malaria, and P. vivax being a second important cause, especially in Asia, the Americas and the Pacific. During infection with Plasmodium spp., the merozoite form of the parasite invades red blood cells and replicates inside them. It is during the blood-stage of infection that malaria disease occurs and, therefore, understanding merozoite invasion, host immune responses to merozoite surface antigens, and targeting merozoite surface proteins and invasion ligands by novel vaccines and therapeutics have been important areas of research. Merozoite invasion involves multiple interactions and events, and substantial processing of merozoite surface proteins occurs before, during and after invasion. The merozoite surface is highly complex, presenting a multitude of antigens to the immune system. This complexity has proved challenging to our efforts to understand merozoite invasion and malaria immunity, and to developing merozoite antigens as malaria vaccines. In recent years, there has been major progress in this field, and several merozoite surface proteins show strong potential as malaria vaccines. Our current knowledge on this topic is reviewed, highlighting recent advances and research priorities. The authors summarize current knowledge of merozoite surface proteins of malaria parasites; their function in invasion, processing of surface proteins before, during and after invasion, their importance as targets of immunity, and the current status of malaria vaccines that target merozoite surface proteins.
Collapse
Affiliation(s)
- James G Beeson
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, Australia Department of Microbiology, Monash University, Clayton, Victoria, Australia Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Damien R Drew
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, Australia
| | - Michelle J Boyle
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, Australia
| | - Gaoqian Feng
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, Australia
| | - Freya J I Fowkes
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, Australia Department of Epidemiology and Preventive Medicine, Monash University, Clayton, Victoria, Australia School of Population Health, University of Melbourne, Parkville, Victoria, Australia
| | - Jack S Richards
- Burnet Institute for Medical Research and Public Health, 85 Commercial Road, Melbourne, Victoria, Australia Department of Microbiology, Monash University, Clayton, Victoria, Australia Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
39
|
Zhu X, Zhao Z, Feng Y, Li P, Liu F, Liu J, Yang Z, Yan G, Fan Q, Cao Y, Cui L. Genetic diversity of the Plasmodium falciparum apical membrane antigen I gene in parasite population from the China-Myanmar border area. INFECTION GENETICS AND EVOLUTION 2016; 39:155-162. [PMID: 26825252 DOI: 10.1016/j.meegid.2016.01.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 01/20/2016] [Accepted: 01/23/2016] [Indexed: 11/28/2022]
Abstract
To investigate the genetic diversity of the Plasmodium falciparum apical membrane antigen 1 (PfAMA1) gene in Southeast Asia, we determined PfAMA1 sequences from 135 field isolates collected from the China-Myanmar border area and compared them with 956 publically available PfAMA1 sequences from seven global P. falciparum populations. This analysis revealed high genetic diversity of PfAMA1 in global P. falciparum populations with a total of 229 haplotypes identified. The genetic diversity of PfAMA1 gene from the China-Myanmar border is not evenly distributed in the different domains of this gene. Sequence diversity in PfAMA1 from the China-Myanmar border is lower than that observed in Thai, African and Oceanian populations, but higher than that in the South American population. This appeared to correlate well with the levels of endemicity of different malaria-endemic regions, where hyperendemic regions favor genetic cross of the parasite isolates and generation of higher genetic diversity. Neutrality tests show significant departure from neutrality in the entire ectodomain and Domain I of PfAMA1 in the China-Myanmar border parasite population. We found evidence supporting a substantial continent-wise genetic structure among P. falciparum populations, with the highest genetic differentiation detected between the China-Myanmar border and the South American populations. Whereas no alleles were unique to a specific region, there were considerable geographical differences in major alleles and their frequencies, highlighting further necessity to include more PfAMA1 alleles in vaccine designs.
Collapse
Affiliation(s)
- Xiaotong Zhu
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning 110122, China
| | - Zhenjun Zhao
- Dalian Institute of Biotechnology, Dalian, Liaoning, China
| | - Yonghui Feng
- Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Peipei Li
- Dalian Institute of Biotechnology, Dalian, Liaoning, China
| | - Fei Liu
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning 110122, China
| | - Jun Liu
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning 110122, China
| | - Zhaoqing Yang
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, China
| | - Guiyun Yan
- Program in Public Health, University of California, Irvine, CA, USA
| | - Qi Fan
- Dalian Institute of Biotechnology, Dalian, Liaoning, China
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning 110122, China.
| | - Liwang Cui
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning 110122, China; Department of Entomology, The Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
40
|
González-Cerón L, Cerritos R, Corzo-Mancilla J, Santillán F. Diversity and evolutionary genetics of the three major Plasmodium vivax merozoite genes participating in reticulocyte invasion in southern Mexico. Parasit Vectors 2015; 8:651. [PMID: 26691669 PMCID: PMC4687067 DOI: 10.1186/s13071-015-1266-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 12/11/2015] [Indexed: 11/25/2022] Open
Abstract
Background Reported malaria cases in the Americas had been reduced to about one-half million by 2012. To advance towards elimination of this disease, it is necessary to gain insights into how the malaria parasite is evolving, including the emergence, spread and persistence of new haplotypes in affected regions. In here, the genetic diversity of the three major P. vivax merozoite genes was analyzed. Methods From P. vivax-infected blood samples obtained in southern Mexico (SMX) during 2006–2007, nucleotide sequences were achieved for: the 42 kDa carboxyl fragment of the merozoite surface protein-1 (msp142), domains I-II of the apical membrane antigen-1 (ama1I-II), and domain II of the Duffy binding protein (dbpII). Gene polymorphism was examined and haplotype networks were developed to depict parasite relationships in SMX. Then genetic diversity, recombination and natural selection were analyzed and the degree of differentiation was determined as FST values. Results The diversity of P. vivax merozoite genes in SMX was less than that of parasites from other geographic origins, with dbpII < ama1I-II < msp142. Ama1I-II and msp142 exposed the more numerous haplotypes exclusive to SMX. While, all dbpII haplotypes from SMX were separated from one to three mutational steps, the networks of ama1I-II and msp142 were more complex; loops and numerous mutational steps were evidenced, likely due to recombination. Sings of local diversification were more evident for msp142. Sixteen combined haplotypes were determined; one of these haplotypes not detected in 2006 was highly frequent in 2007. The Rm value was higher for msp142than for ama1I-II, being insignificant for dbpII. The dN-dS value was highly significant for ama1I-II and lesser so for dbpII. The FST values were higher for dbpII than msp142, and very low for ama1I-II. Conclusions In SMX, P. vivax ama1I-II, dbpII and msp142 demonstrated limited diversity, and exhibited a differentiated parasite population. The results suggest that differential intensities of selective forces are operating on these gene fragments, and probably related to their timing, length of exposure and function during reticulocyte adhesion and invasion. Therefore, these finding are essential for mono and multivalent vaccine development and for epidemiological surveillance. Electronic supplementary material The online version of this article (doi:10.1186/s13071-015-1266-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lilia González-Cerón
- Regional Centre for Research in Public Health, National Institute for Public Health, Tapachula, Chiapas, 30700, Mexico.
| | - Rene Cerritos
- Departamento de Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, México, DF, 04510, México.
| | - Jordán Corzo-Mancilla
- Regional Centre for Research in Public Health, National Institute for Public Health, Tapachula, Chiapas, 30700, Mexico.
| | - Frida Santillán
- Regional Centre for Research in Public Health, National Institute for Public Health, Tapachula, Chiapas, 30700, Mexico.
| |
Collapse
|
41
|
Abstract
In 2013 there were an estimated 584,000 deaths and 198 million clinical illnesses due to malaria, the majority in sub-Saharan Africa. Vaccines would be the ideal addition to the existing armamentarium of anti-malaria tools. However, malaria is caused by parasites, and parasites are much more complex in terms of their biology than the viruses and bacteria for which we have vaccines, passing through multiple stages of development in the human host, each stage expressing hundreds of unique antigens. This complexity makes it more difficult to develop a vaccine for parasites than for viruses and bacteria, since an immune response targeting one stage may not offer protection against a later stage, because different antigens are the targets of protective immunity at different stages. Furthermore, depending on the life cycle stage and whether the parasite is extra- or intra-cellular, antibody and/or cellular immune responses provide protection. It is thus not surprising that there is no vaccine on the market for prevention of malaria, or any human parasitic infection. In fact, no vaccine for any disease with this breadth of targets and immune responses exists. In this limited review, we focus on four approaches to malaria vaccines, (1) a recombinant protein with adjuvant vaccine aimed at Plasmodium falciparum (Pf) pre-erythrocytic stages of the parasite cycle (RTS,S/AS01), (2) whole sporozoite vaccines aimed at Pf pre-erythrocytic stages (PfSPZ Vaccine and PfSPZ-CVac), (3) prime boost vaccines that include recombinant DNA, viruses and bacteria, and protein with adjuvant aimed primarily at Pf pre-erythrocytic, but also asexual erythrocytic stages, and (4) recombinant protein with adjuvant vaccines aimed at Pf and Plasmodium vivax sexual erythrocytic and mosquito stages. We recognize that we are not covering all approaches to malaria vaccine development, or most of the critically important work on development of vaccines against P. vivax, the second most important cause of malaria. Progress during the last few years has been significant, and a first generation malaria candidate vaccine, RTS,S/AS01, is under review by the European Medicines Agency (EMA) for its quality, safety and efficacy under article 58, which allows the EMA to give a scientific opinion about products intended exclusively for markets outside of the European Union. However, much work is in progress to optimize malaria vaccines in regard to magnitude and durability of protective efficacy and the financing and practicality of delivery. Thus, we are hopeful that anti-malaria vaccines will soon be important tools in the battle against malaria.
Collapse
|
42
|
Clinical Variation of Plasmodium falciparum eba-175, ama-1, and msp-3 Genotypes in Young Children Living in a Seasonally High Malaria Transmission Setting in Burkina Faso. J Parasitol Res 2015; 2015:985651. [PMID: 26634149 PMCID: PMC4655070 DOI: 10.1155/2015/985651] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 10/08/2015] [Accepted: 10/15/2015] [Indexed: 11/17/2022] Open
Abstract
The association between P. falciparum eba-175, ama-1, and msp-3 polymorphism in the pathogenicity of malaria disease was investigated. We therefore compared the prevalence of different alleles between symptomatic and asymptomatic malarial children under five years of age living in Burkina Faso. Blood filter papers were collected during the 2008 malaria transmission season from 228 symptomatic and 199 asymptomatic children under five years of age. All patients were living in the rural area of Saponé at about 50 km from Ouagadougou, the capital city of Burkina Faso. P. falciparum parasite DNA was extracted using QIAGEN kits and the alleles diversity was assessed by a nested PCR. PCR products were then digested by restriction enzymes based on already described polymorphic regions of the eba-175, ama-1, and msp-3 genes. The individual alleles eba-175_FCR3 and msp-3_K1 frequencies were statistically higher (p < 0.0001) in the asymptomatic group compared to the symptomatic ones. No statistically significant difference was noted in the prevalence of ama-1-3D7, ama-1-K1, and ama-1-HB3 genotypes between the two groups (p > 0.05). The comparative analysis of P. falciparum genotypes indicated that the polymorphism in eba-175 and msp-3 genotypes varied between asymptomatic and symptomatic clinical groups and may contribute to the pathogenesis of malaria.
Collapse
|
43
|
Designing malaria vaccines to circumvent antigen variability. Vaccine 2015; 33:7506-12. [PMID: 26475447 PMCID: PMC4731100 DOI: 10.1016/j.vaccine.2015.09.110] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 09/29/2015] [Accepted: 09/30/2015] [Indexed: 12/12/2022]
Abstract
Prospects for malaria eradication will be greatly enhanced by an effective vaccine, but parasite genetic diversity poses a major impediment to malaria vaccine efficacy. In recent pre-clinical and field trials, vaccines based on polymorphic Plasmodium falciparum antigens have shown efficacy only against homologous strains, raising the specter of allele-specific immunity such as that which plagues vaccines against influenza and HIV. The most advanced malaria vaccine, RTS,S, targets relatively conserved epitopes on the P. falciparum circumsporozoite protein. After more than 40 years of development and testing, RTS,S, has shown significant but modest efficacy against clinical malaria in phase 2 and 3 trials. Ongoing phase 2 studies of an irradiated sporozoite vaccine will ascertain whether the full protection against homologous experimental malaria challenge conferred by high doses of a whole organism vaccine can provide protection against diverse strains in the field. Here we review and evaluate approaches being taken to design broadly cross-protective malaria vaccines.
Collapse
|
44
|
Draper SJ, Angov E, Horii T, Miller LH, Srinivasan P, Theisen M, Biswas S. Recent advances in recombinant protein-based malaria vaccines. Vaccine 2015; 33:7433-43. [PMID: 26458807 PMCID: PMC4687528 DOI: 10.1016/j.vaccine.2015.09.093] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 09/05/2015] [Accepted: 09/28/2015] [Indexed: 01/03/2023]
Abstract
Protein-based vaccines remain the cornerstone approach for B cell and antibody induction against leading target malaria antigens. Advances in antigen selection, immunogen design and epitope-focusing are advancing the field. New heterologous expression platforms are enabling cGMP production of next-generation protein vaccines. Next-generation antigens, protein-based immunogens and virus-like particle (VLP) delivery platforms are in clinical development. Protein-based vaccines will form part of a highly effective multi-component/multi-stage/multi-antigen subunit formulation against malaria.
Plasmodium parasites are the causative agent of human malaria, and the development of a highly effective vaccine against infection, disease and transmission remains a key priority. It is widely established that multiple stages of the parasite's complex lifecycle within the human host and mosquito vector are susceptible to vaccine-induced antibodies. The mainstay approach to antibody induction by subunit vaccination has been the delivery of protein antigen formulated in adjuvant. Extensive efforts have been made in this endeavor with respect to malaria vaccine development, especially with regard to target antigen discovery, protein expression platforms, adjuvant testing, and development of soluble and virus-like particle (VLP) delivery platforms. The breadth of approaches to protein-based vaccines is continuing to expand as innovative new concepts in next-generation subunit design are explored, with the prospects for the development of a highly effective multi-component/multi-stage/multi-antigen formulation seeming ever more likely. This review will focus on recent progress in protein vaccine design, development and/or clinical testing for a number of leading malaria antigens from the sporozoite-, merozoite- and sexual-stages of the parasite's lifecycle–including PfCelTOS, PfMSP1, PfAMA1, PfRH5, PfSERA5, PfGLURP, PfMSP3, Pfs48/45 and Pfs25. Future prospects and challenges for the development, production, human delivery and assessment of protein-based malaria vaccines are discussed.
Collapse
Affiliation(s)
- Simon J Draper
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK.
| | - Evelina Angov
- Walter Reed Army Institute of Research, U. S. Military Malaria Research Program, Malaria Vaccine Branch, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
| | - Toshihiro Horii
- Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 561-873, Japan
| | - Louis H Miller
- Malaria Cell Biology Section, Laboratory of Malaria and Vector Research, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Prakash Srinivasan
- Malaria Cell Biology Section, Laboratory of Malaria and Vector Research, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark; Centre for Medical Parasitology at Department of International Health, Immunology, and Microbiology and Department of Infectious Diseases, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sumi Biswas
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| |
Collapse
|
45
|
Hoffman SL, Vekemans J, Richie TL, Duffy PE. The march toward malaria vaccines. Vaccine 2015; 33 Suppl 4:D13-23. [PMID: 26324116 DOI: 10.1016/j.vaccine.2015.07.091] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 07/25/2015] [Accepted: 07/27/2015] [Indexed: 01/14/2023]
Abstract
In 2013 there were an estimated 584,000 deaths and 198 million clinical illnesses due to malaria, the majority in sub-Saharan Africa. Vaccines would be the ideal addition to the existing armamentarium of anti-malaria tools. However, malaria is caused by parasites, and parasites are much more complex in terms of their biology than the viruses and bacteria for which we have vaccines, passing through multiple stages of development in the human host, each stage expressing hundreds of unique antigens. This complexity makes it more difficult to develop a vaccine for parasites than for viruses and bacteria, since an immune response targeting one stage may not offer protection against a later stage, because different antigens are the targets of protective immunity at different stages. Furthermore, depending on the life cycle stage and whether the parasite is extra- or intra-cellular, antibody and/or cellular immune responses provide protection. It is thus not surprising that there is no vaccine on the market for prevention of malaria, or any human parasitic infection. In fact, no vaccine for any disease with this breadth of targets and immune responses exists. In this limited review, we focus on four approaches to malaria vaccines, (1) a recombinant protein with adjuvant vaccine aimed at Plasmodium falciparum (Pf) pre-erythrocytic stages of the parasite cycle (RTS,S/AS01), (2) whole sporozoite vaccines aimed at Pf pre-erythrocytic stages (PfSPZ Vaccine and PfSPZ-CVac), (3) prime boost vaccines that include recombinant DNA, viruses and bacteria, and protein with adjuvant aimed primarily at Pf pre-erythrocytic, but also asexual erythrocytic stages, and (4) recombinant protein with adjuvant vaccines aimed at Pf and Plasmodium vivax sexual erythrocytic and mosquito stages. We recognize that we are not covering all approaches to malaria vaccine development, or most of the critically important work on development of vaccines against P. vivax, the second most important cause of malaria. Progress during the last few years has been significant, and a first generation malaria candidate vaccine, RTS,S/AS01, is under review by the European Medicines Agency (EMA) for its quality, safety and efficacy under article 58, which allows the EMA to give a scientific opinion about products intended exclusively for markets outside of the European Union. However, much work is in progress to optimize malaria vaccines in regard to magnitude and durability of protective efficacy and the financing and practicality of delivery. Thus, we are hopeful that anti-malaria vaccines will soon be important tools in the battle against malaria.
Collapse
Affiliation(s)
| | | | | | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| |
Collapse
|
46
|
Tanner M, Greenwood B, Whitty CJM, Ansah EK, Price RN, Dondorp AM, von Seidlein L, Baird JK, Beeson JG, Fowkes FJI, Hemingway J, Marsh K, Osier F. Malaria eradication and elimination: views on how to translate a vision into reality. BMC Med 2015; 13:167. [PMID: 26208740 PMCID: PMC4514994 DOI: 10.1186/s12916-015-0384-6] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Although global efforts in the past decade have halved the number of deaths due to malaria, there are still an estimated 219 million cases of malaria a year, causing more than half a million deaths. In this forum article, we asked experts working in malaria research and control to discuss the ways in which malaria might eventually be eradicated. Their collective views highlight the challenges and opportunities, and explain how multi-factorial and integrated processes could eventually make malaria eradication a reality.
Collapse
Affiliation(s)
- Marcel Tanner
- Swiss Tropical & Public Health Institute, 4002, Basel, Switzerland. .,University of Basel, Basel, Switzerland.
| | - Brian Greenwood
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK.
| | - Christopher J M Whitty
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK.
| | - Evelyn K Ansah
- Research and Development Division, Ghana Health Service, Accra, Ghana.
| | - Ric N Price
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Australia. .,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Arjen M Dondorp
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK. .,Mahidol Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| | - Lorenz von Seidlein
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK. .,Mahidol Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| | - J Kevin Baird
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK. .,Eijkman-Oxford Clinical Research Unit, Jalan Diponegoro No.69, Jakarta, 10430, Indonesia.
| | - James G Beeson
- Burnet Institute, 85 Commercial Road, Melbourne, Victoria, 3004, Australia. .,Department of Microbiology, Monash University, 19 Innovation Walk, Victoria, 3800, Australia.
| | - Freya J I Fowkes
- Burnet Institute, 85 Commercial Road, Melbourne, Victoria, 3004, Australia. .,Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Australia. .,Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia. .,Department of Infectious Diseases, Monash University, Melbourne, Australia.
| | - Janet Hemingway
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK.
| | - Kevin Marsh
- African Academy of Sciences, Miotoni Road, Miotoni Lane, House No. 8 Karen, P.O. Box 24916-00502, Nairobi, Kenya.
| | - Faith Osier
- KEMRI Centre for Geographic Medicine Research-Coast, Kilifi, Kenya.
| |
Collapse
|
47
|
Impact of malaria preexposure on antiparasite cellular and humoral immune responses after controlled human malaria infection. Infect Immun 2015; 83:2185-96. [PMID: 25776749 DOI: 10.1128/iai.03069-14] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 03/09/2015] [Indexed: 11/20/2022] Open
Abstract
To understand the effect of previous malaria exposure on antiparasite immune responses is important for developing successful immunization strategies. Controlled human malaria infections (CHMIs) using cryopreserved Plasmodium falciparum sporozoites provide a unique opportunity to study differences in acquisition or recall of antimalaria immune responses in individuals from different transmission settings and genetic backgrounds. In this study, we compared antiparasite humoral and cellular immune responses in two cohorts of malaria-naive Dutch volunteers and Tanzanians from an area of low malarial endemicity, who were subjected to the identical CHMI protocol by intradermal injection of P. falciparum sporozoites. Samples from both trials were analyzed in parallel in a single center to ensure direct comparability of immunological outcomes. Within the Tanzanian cohort, we distinguished one group with moderate levels of preexisting antibodies to asexual P. falciparum lysate and another that, based on P. falciparum serology, resembled the malaria-naive Dutch cohort. Positive P. falciparum serology at baseline was associated with a lower parasite density at first detection by quantitative PCR (qPCR) after CHMI than that for Tanzanian volunteers with negative serology. Post-CHMI, both Tanzanian groups showed a stronger increase in anti-P. falciparum antibody titers than Dutch volunteers, indicating similar levels of B-cell memory independent of serology. In contrast to the Dutch, Tanzanians failed to increase P. falciparum-specific in vitro recall gamma interferon (IFN-γ) production after CHMI, and innate IFN-γ responses were lower in P. falciparum lysate-seropositive individuals than in seronegative individuals. In conclusion, positive P. falciparum lysate serology can be used to identify individuals with better parasite control but weaker IFN-γ responses in circulating lymphocytes, which may help to stratify volunteers in future CHMI trials in areas where malaria is endemic.
Collapse
|